1
|
Gong Y, Liu Y, Jiang F, Wang X. Ocular Immune-Related Adverse Events Associated with PD-1 Inhibitors: From Molecular Mechanisms to Clinical Management. Semin Ophthalmol 2025; 40:288-305. [PMID: 39606920 DOI: 10.1080/08820538.2024.2433636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Purpose: To help ophthalmologists and oncologists better understand the ocular irAEs secondary to PD-1 inhibitors , enabling early detection and management of ocular complications.Methods: We reviewed case reports and related literatures on ocular irAEs secondary to PD-1 inhibitors in PubMed, including a total of 70 case reports, summarizing and analyzing the specific conditions of these patients.Results: The most common malignant tumors were melanoma (n = 41; 58.6%) and lung cancer (n = 13; 18.6%). The main PD-1 inhibitors used were pembrolizumab (n = 38; 54.3%) and nivolumab (n = 28; 40%). They may result in various ocular complications, with the most common being uveitis (n = 35; 50%) and myasthenia gravis (n = 13; 18.57%). Adverse events concerning the cornea and the retina were reported in 8 cases each (11.43%). Neuro-ophthalmic adverse events were reported in 6 cases (8.57%). Most of these toxicities responded to topical and systemic steroids. Severe manifestations, however, may require temporary or permanent cessation of PD-1 inhibitors treatment.Conclusions: With the increasing use of PD-1 inhibitors, ophthalmologists need to remain sensitive to the clinical manifestations of adverse events to ensure timely diagnosis and management. To improve their quality of life and reduce mortality, oncologists and ophthalmologists should maintain close cooperation and implement multi-disciplinary treatment.
Collapse
Affiliation(s)
- Yuqi Gong
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yushuai Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | | | - Xinghua Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Qian J, Ruan C, Cai Y, Yi W, Liu J, Xu R. Development and validation of a risk-prediction model for adverse drug reactions in real-world cancer patients treated with anlotinib. Ther Adv Drug Saf 2025; 16:20420986251328687. [PMID: 40151494 PMCID: PMC11946284 DOI: 10.1177/20420986251328687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Background The risk factors related to the adverse drug reactions (ADRs) of anlotinib have been rarely investigated. In addition, a corresponding risk prediction model has not been established in China pertaining to anlotinib-related ADRs. Objectives To manage ADRs more efficiently and improve the prognosis of patients administered anlotinib. Design A retrospective analysis was conducted using the medical records of patients diagnosed with cancer who were administered anlotinib after hospitalization between January 1, 2020, and December 31, 2023. Methods We performed a combination of univariate analysis and multivariate binary logistic regression analysis to identify significant factors that can accurately predict ADRs. Model fitting was performed using forward selection. The accuracy of the prediction model was expressed as the area under the receiver operating characteristic curve (AUC). The final ADR risk model was validated. Results In this study, 300 patients who were administered anlotinib were included. Among them, 238 (79.33%) patients experienced at least one ADR. Diagnosis, combination treatment, distant metastasis, treatment lines, and cumulative dose were independent risk factors for the ADRs of anlotinib. The AUC and the concordance index of the nomogram constructed from the above five factors were 0.790 and 0.789, respectively. The results of the Hosmer-Lemeshow test showed that the model was a good fit (p = 0.811). In addition, the decision curve analysis demonstrated a significantly higher net benefit of the model. The external validation indicated that the prediction nomogram was reliable. Conclusion We developed and validated a simple model to use the ADR risk score in patients who were administered anlotinib. This risk prediction model was well-calibrated and discriminative. It can be used as a reference for clinical decision-making. It has clinical significance for preventing ADRs, improving the prognosis of patients, and providing support for the rational use of drugs.
Collapse
Affiliation(s)
- Jiajia Qian
- Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Cong Ruan
- Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yunyun Cai
- Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Weiyi Yi
- Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jiyong Liu
- Minhang Branch, Fudan University Shanghai Cancer Center, No. 106, Ruili Road, Shanghai 200240, China
- Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rui Xu
- Minhang Branch, Fudan University Shanghai Cancer Center, No. 106, Ruili Road, Shanghai 200240, China
| |
Collapse
|
3
|
Li Y, Lu R, Abuduhailili X, Feng Y. NSUN7 promotes cervical cancer progression through activation of ErbB signaling pathway. Funct Integr Genomics 2025; 25:37. [PMID: 39954044 DOI: 10.1007/s10142-025-01546-9] [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: 09/19/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
We aimed to investigate the role of NSUN7 in the progression of Cervical Cancer through a combination of bioinformatics analysis and cell and animal culture experiments. We comprehensively assessed the expression levels of NSUN7 in the TCGA and CCLE databases, and explored its correlations with clinicopathological features, immune cell infiltration, DNA damage repair gene function, drug sensitivity, and methylation status. The NSUN7 gene was disrupted through lentiviral infection, and the effects on cell proliferation, invasion, and apoptosis were evaluated using CCK-8 assay, Transwell migration assay, and flow cytometry analysis. Gene enrichment analysis wasidentify the biological pathways associated with NSUN7 and cervical cancer development. Additionally, a xenograft model of cervical cancer was established to assess the in vivo inhibitory effect of NSUN7 and its impact on pathway molecules. The results of both in vitro and in vivo experiments confirmed that silencing the NSUN7 gene significantly inhibited the growth, spread, and metastasis of cervical cancer cells, while promoting apoptosis. TUNEL assay and HE staining further verified the apoptotic effect of NSUN7 on tumor tissues, and KEGG enrichment analysis revealed a significant enrichment of NSUN7 in the ErbB pathway. Silencing of NSUN7 resulted in a significant down-regulation of key ErbB pathway proteins (HER2, STAT5, PI3K/p-PI3K) as demonstrated by quantitative real-time PCR and Western blot. These findings suggest that NSUN7 may affect the biological behavior of cervical cancer cells and promote tumor development by activating the ErbB signaling pathway.
Collapse
Affiliation(s)
- Yuxia Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Cancer Hospital Affiliated to Xinjiang Medical University, Xinjiang, China
- Clinical Laboratory Center, Cancer Hospital Affiliated to Xinjiang Medical University, Xinjiang, China
| | - Ruijiao Lu
- Clinical Laboratory Center, Cancer Hospital Affiliated to Xinjiang Medical University, Xinjiang, China
| | - Xieyidai Abuduhailili
- Clinical Laboratory Center, Cancer Hospital Affiliated to Xinjiang Medical University, Xinjiang, China
| | - Yangchun Feng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Cancer Hospital Affiliated to Xinjiang Medical University, Xinjiang, China.
- Clinical Laboratory Center, Cancer Hospital Affiliated to Xinjiang Medical University, Xinjiang, China.
- Clinical Laboratory Center, The First Huizhou Affiliated Hospital of Guangdong Medical University, Guangdong, China.
| |
Collapse
|
4
|
Remiszewski P, Filipek K, Pisklak A, Chmiel P, Rutkowski P, Czarnecka AM. Neoadjuvant or Adjuvant Chemotherapy in Soft-Tissue Sarcoma? Curr Oncol Rep 2025:10.1007/s11912-024-01630-6. [PMID: 40153167 DOI: 10.1007/s11912-024-01630-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 03/30/2025]
Abstract
PURPOSE OF REVIEW The review compares the effectiveness of neoadjuvant(pre-operative, NAC) and adjuvant(post-operative, AC) in Soft Tissue Sarcomas as this topic is controvesial and multiple new studies have been over the years. RECENT FINDINGS Sarculator and other nomograms assess patients with a predicted 10-year OS below 60% who will benefit from perioperative chemotherapy. Further research supports perioperative chemotherapy's role. European guidelines do not recommend anthracycline and ifosfamide (AI) perioperative chemotherapy as a standard treatment for STS of the extremities and trunk. However, some studies show that AI chemotherapy can improve recurrence-free survival (RFS). The EORTC 62,771 trial found that the CYVADIC regimen (doxorubicin, dacarbazine, cyclophosphamide, vincristine) reduced RFS without affecting OS. Meanwhile, the EORTC 62,931 trial showed no effect of AI chemotherapy on RFS or OS, but a pooled analysis suggested an OS benefit for patients with R1 (microscopically positive) resections. The AI regimen shows further support from Sarculator-based data, with EORTC 62,931 analysis indicating an improvement in disease-free survival and OS in patients with low expected OS. Similar outcomes were seen in the ISG-STS 1001 study. Recently, PERSARC analysis revealed that AI chemotherapy significantly improves OS in high-grade STS patients with a low 5-year OS prediction (< 33%). NAC improves the chances of complete tumour removal, especially in large, high-grade tumours. It often reduces the need for more aggressive surgeries by shrinking tumours before surgery, leading to higher rates of successful resections with clear margins (R0). Sarculator and other nomograms assess patients with a predicted 10-year OS below 60% who will benefit from perioperative chemotherapy. Further research supports perioperative chemotherapy's role.
Collapse
Affiliation(s)
- Piotr Remiszewski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Medical Faculty, Medical University of Warsaw, Warsaw, Poland
| | - Kinga Filipek
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Regional Hospital in Miedzylesie, Warsaw, 04-749, Poland
| | - Agata Pisklak
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Regional Hospital in Miedzylesie, Warsaw, 04-749, Poland
| | - Paulina Chmiel
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Medical Faculty, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Anna M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
5
|
Colombo A, Concetta PM, Gebbia V, Sambataro D, Scandurra G, Valerio MR. A Narrative Review of the Role of Immunotherapy in Metastatic Carcinoma of the Colon Harboring a BRAF Mutation. In Vivo 2025; 39:25-36. [PMID: 39740863 PMCID: PMC11705148 DOI: 10.21873/invivo.13802] [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: 09/04/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 01/02/2025]
Abstract
Patients affected by metastatic carcinoma of the colon/rectum (mCRC) harboring mutations in the BRAF gene (MBRAF) respond poorly to conventional therapy and have a prognosis worse than that of patients without mutations. Despite the promising outcomes of targeted therapy utilizing multi-targeted inhibition of the mitogen-activated protein kinase (MAPK) signaling system, the therapeutic efficacy, especially for the microsatellite stable/DNA proficient mismatch repair (MSS/PMMR) subtype, remains inadequate. Patients with MBRAF/mCRC and high microsatellite instability or DNA deficient mismatch repair (MSI-H/DMMR) exhibit a substantial tumor mutation burden, suggesting a high probability of response to immunotherapy. It is widely acknowledged that MSS/pMMR/mCRC is an immunologically "cold" malignancy that exhibits resistance to immunotherapy. The integration of targeted therapy and immunotherapy may enhance clinical outcomes in patients with MBRAF/mCRC. Efforts to enhance outcomes are exclusively focused on MSS/DMMR-BRAF mutant cancers, which constitute the largest proportion. This review evaluates the clinical efficacy and advancement of novel immune checkpoint blockade therapies for MSI-H/DMMR and MSS/PMMR BRAF mutant mCRC. We examine potential indicators in the tumor immune milieu for forecasting immunotherapeutic response in BRAF mutant mCRC.
Collapse
Affiliation(s)
| | | | - Vittorio Gebbia
- Medical Oncology, Department of Medicine and Surgery, Kore University of Enna, Enna, Italy;
- Medical Oncology Unit, CdC Torina, Palermo, Italy
| | - Daniela Sambataro
- Medical Oncology, Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Medical Oncology Unit, Ospedale Umberto I, Enna, Italy
| | - Giuseppina Scandurra
- Medical Oncology, Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Medical Oncology Unit, Ospedale Cannizzario, Catania, Italy
| | | |
Collapse
|
6
|
Chowdhury D, Das A, Mishra M, Khutere T, Bodakhe SH. Physiological markers for immunotherapeutics: a review. J Chemother 2024:1-24. [PMID: 39711144 DOI: 10.1080/1120009x.2024.2443701] [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: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Immunotherapy has been advanced through multiple approaches, including immunogenic cytokines, monoclonal antibodies, therapeutic vaccinations, adoptive cell transfer, stem cell transplantation, and oncolytic viruses. This review analyses various strategies in genomics, transcriptomics, single-cell techniques, computational analysis, big data, and imaging technologies for the identification of tumour microbiota and microenvironments. Immunotherapy is becoming acknowledged as a feasible cancer treatment method, facilitating innovative cancer medicines and personalized medicine techniques.
Collapse
Affiliation(s)
- Durlav Chowdhury
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ashmita Das
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Mrityunjay Mishra
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Trinkal Khutere
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
7
|
Ilieva N, Pencheva M, Hadzhiev H, Tashkova D, Daskalova E, Georgiev P, Genova S. Impact of Neoadjuvant Therapy on PD-L1 Expression in Triple-Negative Breast Cancer and Correlation with Clinicopathological Factors. Diagnostics (Basel) 2024; 14:2672. [PMID: 39682581 DOI: 10.3390/diagnostics14232672] [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: 10/17/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND This study aims to deliver more insights on the impact of neoadjuvant treatment on Pd-L1 expression and to evaluate its correlation with clinicopathological factors. METHODS We reviewed 88 TNBC cases for the period 2021-2023. Data on age, tumor size, stage, and treatment were collected. Histological slides were assessed for subtype, grade, and TILs. A total of 48 received neoadjuvant treatment. HER2 and Ki67 were evaluated via immunohistochemistry. PD-L1 expression was tested on primary and residual tumors. Statistical analysis was performed using IBM SPSS (p < 0.05). RESULTS In this study, PD-L1 positive expression was found in 44.3% of primary tumors, with 52.9% of initially positive cases losing expression post-treatment. TILs were significantly higher in PD-L1-positive tumors (mean 41.79% vs. 27.55%, p = 0.001). A notable correlation was found between PD-L1 expression and Ki-67 proliferation index, with PD-L1-positive tumors having a median Ki-67 of 64.49 compared to 52.86 in negative cases (p = 0.015). Neoadjuvant immunotherapy led to a lower mean residual cancer burden (0.95 vs. 2.55, p = 0.002) compared to chemotherapy alone. Higher Ki-67 levels (≥50%) were associated with better treatment outcomes, showing a mean RCB score of 1.60 versus 3.16 for lower levels (p = 0.022). HER2-negative cases had a higher prevalence of favorable pathological response (54.5%) compared to HER2-low tumors (25%, p = 0.048), because of the strong correlation to high proliferative index. CONCLUSIONS In conclusion, PD-L1 expression in TNBC shows significant discordance post-treatment, highlighting the need for routine testing and further research on predictive biomarkers.
Collapse
Affiliation(s)
- Nevena Ilieva
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
- Clinical Pathology Department, Complex Oncology Center Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Mina Pencheva
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Hristo Hadzhiev
- First Oncological Department, Complex Oncology Center Plovdiv, Bul. Al. Stamboliyski 2A, 4000 Plovdiv, Bulgaria
| | - Desislava Tashkova
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
- Clinical Pathology Department, Complex Oncology Center Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Elena Daskalova
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Petar Georgiev
- Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Sylvia Genova
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| |
Collapse
|
8
|
Isono T, Furuno H, Onodera Y, Maruyama T, Takeuchi Y, Ayaka Kojima, Nishida T, Kobayashi Y, Ishiguro T, Takaku Y, Kurashima K, Kagiyama N. Analysis of immune checkpoint inhibitors for advanced non-small cell lung cancer in patients receiving antacids. Respir Investig 2024; 62:951-959. [PMID: 39186878 DOI: 10.1016/j.resinv.2024.08.013] [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: 06/12/2024] [Revised: 08/08/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are reported to decrease the efficacy of immune checkpoint inhibitors (ICIs), but there are few reports on the association between ICI efficacy and antacids other than PPIs, and simultaneous examination of the effects of antacids, corticosteroids, and non-steroidal anti-inflammatory drugs (NSAIDs) on ICI therapy. METHODS We conducted a retrospective study of 381 patients with non-small cell lung cancer who received ICI therapy from January 1, 2016 to December 31, 2022. The primary endpoint was overall survival (OS) and the secondary endpoint was progression-free survival (PFS). Antacids included histamine type 2 receptor antagonists (H2RAs), PPIs, and potassium-competitive acid blockers (P-CABs). RESULTS Antacids were administered to 218 patients, including 168 with PPIs, 37 with P-CABs, and 13 with H2RAs. Patients with antacids had worse median PFS and OS than those without antacids (PFS, 2.9 vs. 6.2 months; OS, 12.3 vs. 24.0 months), and those with PPIs, P-CABs, or H2RAs had similar results. However, there were no significant differences between patients with and without antacids when stratified by corticosteroid and NSAID use. Multivariate analyses showed that corticosteroids and NSAIDs administered for cancer-associated symptoms were related to poor prognosis, but antacids including PPIs, P-CABs, or H2RAs were not related. CONCLUSIONS Antacids were not related to ICI efficacy when NSAIDs or corticosteroids were taken into consideration. This may be because the most frequent reason for administering NSAIDs and corticosteroids was cancer-associated symptoms, which are a poor prognostic factor, and most of the patients treated with these medications also received antacids.
Collapse
Affiliation(s)
- Taisuke Isono
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan.
| | - Hajime Furuno
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Yoko Onodera
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Tomoya Maruyama
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Yuki Takeuchi
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Ayaka Kojima
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Takashi Nishida
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Yoichi Kobayashi
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Takashi Ishiguro
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Yotaro Takaku
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Kazuyoshi Kurashima
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| | - Naho Kagiyama
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, 1696 Itai, Kumagaya, Saitama, 360-0197, Japan
| |
Collapse
|
9
|
Asher N, Bar-Hai N, Ben-Betzalel G, Stoff R, Grynberg S, Schachter J, Frommer-Shapira R. Exploring the clinical significance of specific immune-related adverse events in melanoma patients undergoing immune checkpoint inhibitor therapy. Melanoma Res 2024; 34:439-449. [PMID: 38913412 DOI: 10.1097/cmr.0000000000000985] [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: 06/25/2024]
Abstract
Several studies have demonstrated that patients who experience immune-related adverse events (irAE) as a result of immunotherapy treatment, exhibit significantly improved outcomes compared to patients without toxicity. Data regarding the impact of specific irAE is, however, currently lacking. This is a real-world single-site cohort of 415 advanced melanoma patients who were treated with immunotherapy as first-line between 2014 and 2020, with a median follow-up of 24.5 months. The most frequent irAEs were cutaneous (classified as non-vitiligo, n = 110, 26.5% and vitiligo, n = 48, 11.6%), rheumatologic ( n = 68, 16.4%), gastrointestinal ( n = 66, 15.9%), endocrine ( n = 61, 14.7%), and hepatitis ( n = 50, 12%). Specific irAE that were significantly associated with survival benefit were rheumatologic (hazard ratio 0.34 for PFS, P < 0.001; hazard ratio 0.38 for OS, P < 0.001), non-vitiligo cutaneous (hazard ratio 0.58 for PFS, P < 0.001; hazard ratio 0.54 for OS, P = 0.001), vitiligo (hazard ratio 0.30 for PFS, P < 0.001; hazard ratio 0.29 for OS, P < 0.001), and endocrine (hazard ratio 0.6 for PFS, P = 0.01; hazard ratio 0.52 for OS, P < 0.001). Other types of irAEs, such as colitis, hepatitis and others - do not present this correlation. The occurrence of these specific irAEs may reflect a hyperactivated immune response and thus can serve as meaningful clinical biomarkers.
Collapse
Affiliation(s)
- Nethanel Asher
- Skin Cancer and Melanoma Center, Davidoff Cancer Center, Beilinson Medical Center, Petah Tikva
| | - Neta Bar-Hai
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-Hashomer, Ramat Gan
| | - Guy Ben-Betzalel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-Hashomer, Ramat Gan
| | - Ronen Stoff
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-Hashomer, Ramat Gan
| | - Shirly Grynberg
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-Hashomer, Ramat Gan
| | - Jacob Schachter
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-Hashomer, Ramat Gan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Ronnie Frommer-Shapira
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-Hashomer, Ramat Gan
| |
Collapse
|
10
|
Qian YL, Jiang Y, Gong YH, Yu JK, Liu C, Wu WT, Shen D. Autoimmune encephalitis following treatment with durvalumab for small-cell lung cancer. J Int Med Res 2024; 52:3000605241287015. [PMID: 39435557 PMCID: PMC11497526 DOI: 10.1177/03000605241287015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/10/2024] [Indexed: 10/23/2024] Open
Abstract
The traditional treatment for small-cell lung cancer (SCLC) has been traditional systemic platinum-containing chemotherapy because the response rate is 50-90%. Durvalumab is an immune checkpoint inhibitor that blocks the binding of programmed cell death protein 1 and programmed cell death 1 ligand 1. Durvalumab combined with traditional chemotherapy agents has been recommended as the first-line treatment for extensive-stage SCLC, but its use may cause immune-related adverse events. Autoimmune encephalitis is a rare and potentially fatal neurological adverse event. This current case report describes a male patient in his late 50s with ES-SCLC who developed autoimmune encephalitis associated with durvalumab treatment after three cycles of combination chemotherapy. This current case furthers the understanding of autoimmune encephalitis caused by durvalumab treatment.
Collapse
Affiliation(s)
- Yu-Lan Qian
- Department of Pharmacy, The First affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ye Jiang
- Department of Pharmacy, The First affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Department of Pharmacy, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu Province, China
| | - Yin-Hua Gong
- Department of Pharmacy, The First affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jian-Kang Yu
- Department of Pharmacy, The First affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Chao Liu
- Department of Pulmonary and Critical Care Medicine, The First affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Wen-Ting Wu
- Department of Pulmonary and Critical Care Medicine, The First affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Dan Shen
- Department of Pulmonary and Critical Care Medicine, The First affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
11
|
Fujii T, Kuzuya T, Kondo N, Funasaka K, Ohno E, Hirooka Y, Tochio T. Altered intestinal Streptococcus anginosus and 5α-reductase gene levels in patients with hepatocellular carcinoma and elevated Bacteroides stercoris in atezolizumab/bevacizumab non-responders. J Med Microbiol 2024; 73. [PMID: 39240069 DOI: 10.1099/jmm.0.001878] [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: 09/07/2024] Open
Abstract
Introduction. Hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide.Gap statement. Monitoring of HCC and predicting its immunotherapy responses are challenging.Aim. This study explored the potential of the gut microbiome for HCC monitoring and predicting HCC immunotherapy responses.Methods. DNA samples were collected from the faeces of 22 patients with HCC treated with atezolizumab/bevacizumab (Atz/Bev) and 85 healthy controls. The gut microbiome was analysed using 16S rRNA next-generation sequencing and quantitative PCR (qPCR).Results. The microbiomes of patients with HCC demonstrated significant enrichment of Lactobacillus, particularly Lactobacillus fermentum, and Streptococcus, notably Streptococcus anginosus. Comparative analysis between Atz/Bev responders (R) and non-responders (NR) revealed a higher abundance of Bacteroides stercoris in the NR group and Bacteroides coprocola in the R group. Using qPCR analysis, we observed elevated levels of S. anginosus and reduced levels of 5α-reductase genes, essential for the synthesis of isoallolithocholic acid, in HCC patients compared to controls. Additionally, the analysis confirmed a significantly lower abundance of B. stercoris in the Atz/Bev R group relative to the NR group.Conclusions. The gut microbiome analysis and specific gene quantification via qPCR could provide a rapid, less invasive, and cost-effective approach for assessing the increased risk of HCC, monitoring patient status, and predicting immunotherapy responses.
Collapse
Affiliation(s)
- Tadashi Fujii
- Department of Gastroenterology and Hepatology, Fujita Health University, Aichi, Japan
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Aichi, Japan
- BIOSIS Lab. Co. Ltd., Aichi, Japan
| | - Teiji Kuzuya
- Department of Gastroenterology and Hepatology, Fujita Health University, Aichi, Japan
| | | | - Kohei Funasaka
- Department of Gastroenterology and Hepatology, Fujita Health University, Aichi, Japan
| | - Eizaburo Ohno
- Department of Gastroenterology and Hepatology, Fujita Health University, Aichi, Japan
| | - Yoshiki Hirooka
- Department of Gastroenterology and Hepatology, Fujita Health University, Aichi, Japan
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Aichi, Japan
- BIOSIS Lab. Co. Ltd., Aichi, Japan
| | - Takumi Tochio
- Department of Gastroenterology and Hepatology, Fujita Health University, Aichi, Japan
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Aichi, Japan
- BIOSIS Lab. Co. Ltd., Aichi, Japan
| |
Collapse
|
12
|
Zhao Q, Wang L, Yang X, Feng J, Chen Q. Association of the Scottish inflammatory prognostic score with treatment-related adverse events and prognosis in esophageal cancer receiving neoadjuvant immunochemotherapy. Front Immunol 2024; 15:1418286. [PMID: 39035002 PMCID: PMC11257864 DOI: 10.3389/fimmu.2024.1418286] [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: 04/16/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
Background To investigate the relationship between the Scottish inflammatory prognostic score (SIPS), treatment-related adverse events (TRAEs), and prognostication in patients with neoadjuvant immunochemotherapy (NICT) for esophageal squamous cell carcinoma (ESCC). Methods A retrospective investigation was carried out on 208 ESCC patients treated with NICT. The relationships between the SIPS, TRAEs, and prognosis [disease-free survival (DFS) and overall survival (OS)] were analyzed. Results The patients, comprising 62 (29.8%) cases of SIPS0, 103 (49.5%) cases of SIPS1, and 43 (20.7%) cases of SIPS2, were categorized into three groups based on SIPS. Among patients with SIPS2, the oldest age (P=0.006), lowest BMI (P=0.001), longest tumor length (P=0.001), most advanced ypT stage (P=0.014), and ypN stage (P<0.001) were identified. Pathological complete response (PCR) rates showed statistically significant variations between the three groups (SIPS0: 45.2%, SIPS1: 27.2%, SIPS2: 16.3%, P=0.004). All TRAEs were found in 63.9% (133 cases) of the cases, with serious TRAEs (grade 3-4) accounting for 13.9% (29 cases). TRAEs themselves were not linked with SIPS (P=0.668), while serious TRAEs had a significant correlation with SIPS (P=0.002). Multivariate logistic analysis showed that SIPS2 seemed to confer serious TRAEs [odds radio (OR)=4.044; 95% CI: 1.395-11.722; P=0.010]. For patients classified as SIPS0, 1, or 2, the 3-year DFS was 83.9%, 58.3%, and 39.5% (P<0.001). The 3-year OS for those with SIPS0, 1, or 2 was 88.7%, 72.8%, and 53.5%, respectively (P<0.001). SIPS was substantially correlated with DFS (but not with OS) and could be utilized as an independent predictor [SIPS2: hazard ratio (HR)=3.743, 95% CI: 1.770-7.914, P=0.001; SIPS1: HR=2.303, 95% CI: 1.149-4.616, P=0.019]. Conclusion The SIPS is associated with serious TRAEs and can be used as a predictor of serious TRAEs in ESCC receiving NICT. SIPS may be employed for pretreatment assessment since it was found to be substantially correlated with DFS.
Collapse
Affiliation(s)
- Qiang Zhao
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus) of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Liang Wang
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xun Yang
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Jifeng Feng
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus) of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Qixun Chen
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus) of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
13
|
Zhao Z, Niu J, Wang J, Zhang R, Liang H, Ma Y, Ferrena A, Wang W, Yang R, Geller DS, Guo W, Ren T, Hoang BH, Tang X, Yan T. Novel candidate metastasis-associated genes for synovial sarcoma. J Cell Mol Med 2024; 28:e18541. [PMID: 39046429 PMCID: PMC11267982 DOI: 10.1111/jcmm.18541] [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: 11/02/2023] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
Synovial sarcoma (SS) is an aggressive soft tissue sarcoma with poor prognosis due to late recurrence and metastasis. Metastasis is an important prognostic factor of SS. This study aimed to identify the core genes and mechanisms associated with SS metastasis. Microarray data for GSE40021 and GSE40018 were obtained from the Gene Expression Omnibus database. 186 differentially expressed genes (DEGs) were identified. The biological functions and signalling pathways closely associated with SS metastasis included extracellular matrix (ECM) organization and ECM-receptor interaction. Gene set enrichment analysis showed that the terms cell cycle, DNA replication, homologous recombination and mismatch repair were significantly enriched in the metastasis group. Weighted gene co-expression network analysis identified the most relevant module and 133 hub genes, and 31 crossover genes were identified by combining DEGs. Subsequently, four characteristic genes, EXO1, NCAPG, POLQ and UHRF1, were identified as potential biomarkers associated with SS metastasis using the least absolute shrinkage and selection operator algorithm and validation dataset verification analysis. Immunohistochemistry results from our cohort of 49 patients revealed visible differences in the expression of characteristic genes between the non-metastatic and metastatic groups. Survival analysis indicated that high expression of characteristic genes predicted poor prognosis. Our data revealed that primary SS samples from patients who developed metastasis showed activated homologous recombination and mismatch repair compared to samples from patients without metastasis. Furthermore, EXO1, NCAPG, POLQ and UHRF1 were identified as potential candidate metastasis-associated genes. This study provides further research insights and helps explore the mechanisms of SS metastasis.
Collapse
Affiliation(s)
- Zhiqing Zhao
- Department of OrthopedicsPeking University First HospitalBeijingChina
| | - Jianfang Niu
- Department of OrthopedicsPeking University First HospitalBeijingChina
| | - Jichuan Wang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
- Beijing Key Laboratory of Musculoskeletal TumorBeijingChina
| | - Ranxin Zhang
- Department of Orthopedic Surgery, Montefiore Medical CenterAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Haijie Liang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
- Beijing Key Laboratory of Musculoskeletal TumorBeijingChina
| | - Yingteng Ma
- Department of PathologyPeking University People's HospitalBeijingChina
| | - Alexander Ferrena
- Department of Orthopedic Surgery, Montefiore Medical CenterAlbert Einstein College of MedicineBronxNew YorkUSA
- Department of Genetics, Institute for Clinical and Translational ResearchAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Wei Wang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
- Beijing Key Laboratory of Musculoskeletal TumorBeijingChina
| | - Rui Yang
- Department of Orthopedic Surgery, Montefiore Medical CenterAlbert Einstein College of MedicineBronxNew YorkUSA
| | - David S. Geller
- Department of Orthopedic Surgery, Montefiore Medical CenterAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Wei Guo
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
- Beijing Key Laboratory of Musculoskeletal TumorBeijingChina
| | - Tingting Ren
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
- Beijing Key Laboratory of Musculoskeletal TumorBeijingChina
| | - Bang H. Hoang
- Department of Orthopedic Surgery, Montefiore Medical CenterAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Xiaodong Tang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
- Beijing Key Laboratory of Musculoskeletal TumorBeijingChina
| | - Taiqiang Yan
- Department of OrthopedicsPeking University First HospitalBeijingChina
| |
Collapse
|
14
|
Emens LA, Romero PJ, Anderson AC, Bruno TC, Capitini CM, Collyar D, Gulley JL, Hwu P, Posey AD, Silk AW, Wargo JA. Challenges and opportunities in cancer immunotherapy: a Society for Immunotherapy of Cancer (SITC) strategic vision. J Immunother Cancer 2024; 12:e009063. [PMID: 38901879 PMCID: PMC11191773 DOI: 10.1136/jitc-2024-009063] [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] [Accepted: 04/24/2024] [Indexed: 06/22/2024] Open
Abstract
Cancer immunotherapy has flourished over the last 10-15 years, transforming the practice of oncology and providing long-term clinical benefit to some patients. During this time, three distinct classes of immune checkpoint inhibitors, chimeric antigen receptor-T cell therapies specific for two targets, and two distinct classes of bispecific T cell engagers, a vaccine, and an oncolytic virus have joined cytokines as a standard of cancer care. At the same time, scientific progress has delivered vast amounts of new knowledge. For example, advances in technologies such as single-cell sequencing and spatial transcriptomics have provided deep insights into the immunobiology of the tumor microenvironment. With this rapid clinical and scientific progress, the field of cancer immunotherapy is currently at a critical inflection point, with potential for exponential growth over the next decade. Recognizing this, the Society for Immunotherapy of Cancer convened a diverse group of experts in cancer immunotherapy representing academia, the pharmaceutical and biotechnology industries, patient advocacy, and the regulatory community to identify current opportunities and challenges with the goal of prioritizing areas with the highest potential for clinical impact. The consensus group identified seven high-priority areas of current opportunity for the field: mechanisms of antitumor activity and toxicity; mechanisms of drug resistance; biomarkers and biospecimens; unique aspects of novel therapeutics; host and environmental interactions; premalignant immunity, immune interception, and immunoprevention; and clinical trial design, endpoints, and conduct. Additionally, potential roadblocks to progress were discussed, and several topics were identified as cross-cutting tools for optimization, each with potential to impact multiple scientific priority areas. These cross-cutting tools include preclinical models, data curation and sharing, biopsies and biospecimens, diversification of funding sources, definitions and standards, and patient engagement. Finally, three key guiding principles were identified that will both optimize and maximize progress in the field. These include engaging the patient community; cultivating diversity, equity, inclusion, and accessibility; and leveraging the power of artificial intelligence to accelerate progress. Here, we present the outcomes of these discussions as a strategic vision to galvanize the field for the next decade of exponential progress in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Ana Carrizosa Anderson
- The Gene Lay Institute of Immunology and Inflammation, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Tullia C Bruno
- Department of Immunology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christian M Capitini
- Department of Pediatrics and Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Deborah Collyar
- Patient Advocates in Research (PAIR), Danville, California, USA
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Avery D Posey
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ann W Silk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
15
|
Oya K, Nakamura Y, Shen LTW, Ishizuki S, Matsusaka S, Fujisawa Y. Soluble PD-L1 predicts tumor response and immune-related adverse events in patients with advanced melanoma treated with anti-PD-1 antibodies. J Dermatol 2024; 51:807-815. [PMID: 38433350 DOI: 10.1111/1346-8138.17183] [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: 12/01/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Immune checkpoint inhibitors (ICIs) bring prognostic benefits to patients with malignancies. However, there is a substantial number of patients whose lesions are not improved by ICIs. In addition, ICIs may cause immune-related adverse events (irAEs), which could lead to an unfavorable prognosis with fatal consequences. Therefore, we conducted a retrospective study to evaluate the utility of circulating sPD-L1 (soluble programmed cell death 1 ligand 1) as a biomarker in patients with advanced melanoma treated with anti-PD-1 (programmed cell death 1 protein) antibodies. Sera from 31 consecutive patients were prospectively collected before and after anti-PD-1 antibody treatment and the serum level of sPD-L1 was evaluated. We found that high sPD-L1 levels before treatment were associated with better prognosis, and this association was observed only in patients with a low tumor burden. We also found that sPD-L1 levels were elevated in patients who developed severe irAEs after treatment, and the patients with severe irAEs had significantly higher fluctuations in sPD-L1 (delta sPD-L1) than those without severe irAEs. Our study suggests that serum sPD-L1 level is a useful biomarker to predict tumor response and irAE development in patients with advanced melanoma treated with anti-PD-1 antibodies.
Collapse
Affiliation(s)
- Kazumasa Oya
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshiyuki Nakamura
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Larina Tzu-Wei Shen
- Department of Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shoichiro Ishizuki
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoshi Matsusaka
- Department of Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Tsukuba Clinical Research and Development Organization, University of Tsukuba, Tsukuba, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Ehime University Graduate School of Medicine, Toon, Japan
| |
Collapse
|
16
|
Sato Y, Nakamura T, Yamada Y, Harashima H. The impact of, and expectations for, lipid nanoparticle technology: From cellular targeting to organelle targeting. J Control Release 2024; 370:516-527. [PMID: 38718875 DOI: 10.1016/j.jconrel.2024.05.006] [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: 02/06/2024] [Revised: 04/22/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
The success of mRNA vaccines against COVID-19 has enhanced the potential of lipid nanoparticles (LNPs) as a system for the delivery of mRNA. In this review, we describe our progress using a lipid library to engineer ionizable lipids and promote LNP technology from the viewpoints of safety, controlled biodistribution, and mRNA vaccines. These advancements in LNP technology are applied to cancer immunology, and a potential nano-DDS is constructed to evaluate immune status that is associated with a cancer-immunity cycle that includes the sub-cycles in tumor microenvironments. We also discuss the importance of the delivery of antigens and adjuvants in enhancing the cancer-immunity cycle. Recent progress in NK cell targeting in cancer immunotherapy is also introduced. Finally, the impact of next-generation DDS technology is explained using the MITO-Porter membrane fusion-based delivery system for the organelle targeting of the mitochondria. We introduce a successful example of the MITO-Porter used in a cell therapeutic strategy to treat cardiomyopathy.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan
| | | |
Collapse
|
17
|
Zhao J, Liu G, Yang X, Zhang C, Han B, Jiang M. Research hot spots and trends in endocrine-related adverse events caused by immune checkpoint inhibitors: a bibliometric analysis and visualization research. Front Endocrinol (Lausanne) 2024; 15:1253832. [PMID: 38686201 PMCID: PMC11056583 DOI: 10.3389/fendo.2024.1253832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/14/2024] [Indexed: 05/02/2024] Open
Abstract
Background In recent years, with the widespread use of immune checkpoint inhibitors (ICIs) in cancer treatment, the toxicity associated with immunotherapy of ICIs has attracted more attention from scholars. Endocrine toxicity is the most likely immune-related adverse events (irAEs) and is often irreversible, posing a significant clinical treatment challenge. Methods In this study, bibliometric methods were used to analyze relevant literature in screening endocrine-related adverse events caused by ICIs in the Web of Science core collection database (WoSCC) and to summarize the status, research hot spots, and future trends in this field. Results 321 countries, 297 institutions, 365 authors, and 305 journals had published 671 English documents on endocrine adverse reactions of ICIs as of 1 December, 2022. The United States, Japan, and China were the top three countries with the most publications. The University of Texas MD Anderson Cancer Center, Harvard Medical School, and Memorial Sloan Kettering Cancer Center were the top three research institutions in terms of publication output. F Stephen Hodi, from the Dana-Farber Cancer Institute in the United States, contributed the largest number of publications. Frontiers in Oncology, which was the most widely distributed publication in the field. The main keywords or clusters identified that current research hotspots include the management of endocrine-related adverse events, hypophysitis, thyroid dysfunction, type I diabetes mellitus, and the impact of endocrine adverse events on survival of patients in this field. Conclusion The basic knowledge structure of the field of endocrine-related adverse events of ICIs, including publication trends, authors, institutions, countries, keywords, journals and publications, and cited documents, was visually analyzed in this bibliometric analysis. The research results comprehensively demonstrated the hot spots and future trends in the research field, as well as its broad prospects, thus providing a reference for the researchers.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guangwei Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xue Yang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chuanzhou Zhang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Bing Han
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Man Jiang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
18
|
Bai R, Yin P, Xing Z, Wu S, Zhang W, Ma X, Gan X, Liang Y, Zang Q, Lei H, Wei Y, Zhang C, Dai B, Zheng Y. Investigation of GPR143 as a promising novel marker for the progression of skin cutaneous melanoma through bioinformatic analyses and cell experiments. Apoptosis 2024; 29:372-392. [PMID: 37945816 DOI: 10.1007/s10495-023-01913-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Skin cutaneous melanoma (SKCM) is an aggressive and life-threatening skin cancer. G-protein coupled receptor 143 (GPR143) belongs to the superfamily of G protein-coupled receptors. METHODS We used the TCGA, GTEx, CCLE, and the Human Protein Atlas databases to examine the mRNA and protein expression of GPR143. In addition, we performed a survival analysis and evaluated the diagnostic efficacy using the Receiver-Operating Characteristic (ROC) curve. Through CIBERSORT, R programming, TIMER, Gene Expression Profiling Interactive Analysis, Sangerbox, and Kaplan-Meier plotter database analyses, we explored the relationships between GPR143, immune infiltration, and gene marker expression of immune infiltrated cells. Furthermore, we investigated the proteins that potentially interact with GPR143 and their functions using R programming and databases including STRING, GeneMANIA, and GSEA. Meanwhile, the cBioPortal, UALCNA, and the MethSurv databases were used to examine the genomic alteration and methylation of GPR143 in SKCM. The Connectivity Map database was used to discover potentially effective therapeutic molecules against SKCM. Finally, we conducted cell experiments to investigate the potential role of GPR143 in SKCM. RESULTS We demonstrated a significantly high expression level of GPR143 in SKCM compared with normal tissues. High GPR143 expression and hypomethylation status of GPR143 were associated with a poorer prognosis. ROC analysis showed that the diagnostic efficacy of the GPR143 was 0.900. Furthermore, GPR143 expression was significantly correlated with immune infiltration in SKCM. We identified 20 neighbor genes and the pathways they enriched were anabolic process of pigmentation, immune regulation, and so on. Genomic alteration analysis revealed significantly different copy number variations related to GPR143 expression in SKCM, and shallow deletion could lead to high expression of GPR143. Ten potential therapeutic drugs against SKCM were identified. GPR143 knockdown inhibited melanoma cell proliferation, migration, and colony formation while promoting apoptosis. CONCLUSIONS Our findings suggest that GPR143 serves as a novel diagnostic and prognostic biomarker and is associated with the progression of SKCM.
Collapse
Affiliation(s)
- Ruimin Bai
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Pan Yin
- Department of Medicine, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Zixuan Xing
- Department of Medicine, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Shaobo Wu
- Department of Medicine, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China.
| | - Wen Zhang
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Xinyu Ma
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Xinyi Gan
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yuxia Liang
- Department of Imaging, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Qijuan Zang
- Department of Medicine, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Hao Lei
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yi Wei
- Department of Urology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Chaonan Zhang
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Bingling Dai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China.
| | - Yan Zheng
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
19
|
Pullen RH, Sassano E, Agrawal P, Escobar J, Chehtane M, Schanen B, Drake DR, Luna E, Brennan RJ. A Predictive Model of Vaccine Reactogenicity Using Data from an In Vitro Human Innate Immunity Assay System. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:904-916. [PMID: 38276072 DOI: 10.4049/jimmunol.2300185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 01/02/2024] [Indexed: 01/27/2024]
Abstract
A primary concern in vaccine development is safety, particularly avoiding an excessive immune reaction in an otherwise healthy individual. An accurate prediction of vaccine reactogenicity using in vitro assays and computational models would facilitate screening and prioritization of novel candidates early in the vaccine development process. Using the modular in vitro immune construct model of human innate immunity, PBMCs from 40 healthy donors were treated with 10 different vaccines of varying reactogenicity profiles and then cell culture supernatants were analyzed via flow cytometry and a multichemokine/cytokine assay. Differential response profiles of innate activity and cell viability were observed in the system. In parallel, an extensive adverse event (AE) dataset for the vaccines was assembled from clinical trial data. A novel reactogenicity scoring framework accounting for the frequency and severity of local and systemic AEs was applied to the clinical data, and a machine learning approach was employed to predict the incidence of clinical AEs from the in vitro assay data. Biomarker analysis suggested that the relative levels of IL-1B, IL-6, IL-10, and CCL4 have higher predictive importance for AE risk. Predictive models were developed for local reactogenicity, systemic reactogenicity, and specific individual AEs. A forward-validation study was performed with a vaccine not used in model development, Trumenba (meningococcal group B vaccine). The clinically observed Trumenba local and systemic reactogenicity fell on the 26th and 93rd percentiles of the ranges predicted by the respective models. Models predicting specific AEs were less accurate. Our study presents a useful framework for the further development of vaccine reactogenicity predictive models.
Collapse
|
20
|
Velikova T, Krastev B, Gulinac M, Zashev M, Graklanov V, Peruhova M. New strategies in the diagnosis and treatment of immune-checkpoint inhibitor-mediated colitis. World J Clin Cases 2024; 12:1050-1062. [PMID: 38464930 PMCID: PMC10921308 DOI: 10.12998/wjcc.v12.i6.1050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/20/2023] [Accepted: 01/19/2024] [Indexed: 02/20/2024] Open
Abstract
Immune-checkpoint inhibitor-mediated colitis (IMC) is an increasingly recognized adverse event in cancer immunotherapy, particularly associated with immune checkpoint inhibitors (ICIs) such as anti-cytotoxic T-lymphocyte antigen-4 and anti-programmed cell death protein-1 antibodies. As this revolutionary immunotherapy gains prominence in cancer treatment, understanding, diagnosing, and effectively managing IMC becomes paramount. IMC represents a unique challenge due to its immune-mediated nature and potential for severe complications. However, a precise picture of IMC pathophysiology is currently unavailable. Therefore, we aimed to summarize the existing data while acknowledging the need for further research. This comprehensive review explores the mechanisms underlying ICIs, gastrointestinal adverse effects, and, in particular, IMC's incidence, prevalence, and features. Our review also emphasizes the importance of recognizing IMC's distinct clinical and histopathological features to differentiate it from other forms of colitis. Furthermore, this paper highlights the urgent need for evolving diagnostic methods, therapeutic strategies, and a multidisciplinary approach to effectively manage IMC.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Boris Krastev
- Medical Center Nadezhda, Medical Center Nadezhda, Sofia 1407, Bulgaria
| | - Milena Gulinac
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
- General and Clinical Pathology, Medical University of Plovdiv, Plovdiv 4002, Bulgaria
| | - Miroslav Zashev
- Department of General Surgery, University Hospital “Heart and Brain”, Burgas 8000, Bulgaria
| | - Vasko Graklanov
- First Department of Internal Diseases, Medical University of Plovdiv, Plovdiv 4000, Bulgaria
- Department of Hematology, University Hospital “St. George”, Plovdiv 4000, Bulgaria
| | - Milena Peruhova
- Division of Gastroenterology, University Hospital “Heart and Brain”, Burgas 1000, Bulgaria
| |
Collapse
|
21
|
QI C, TIAN P, LI W. [Progress on Neoadjuvant Immunotherapy for Resectable Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:138-146. [PMID: 38453446 PMCID: PMC10918244 DOI: 10.3779/j.issn.1009-3419.2024.102.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Indexed: 03/09/2024]
Abstract
In recent years, there has been a consensus regarding the enhancement of prognosis in patients with advanced non-small cell lung cancer (NSCLC) through the utilization of immune checkpoint inhibitors (ICIs). Numerous clinical studies have also demonstrated the substantial value of immunotherapy for resectable NSCLC patients. Nevertheless, there remain controversies surrounding the exploration of immune combination strategies, treatment-related side effects, prognostic biomarkers, as well as other issues in the neoadjuvant therapy setting. Consequently, this article presents a comprehensive overview of the recent advancements in neoadjuvant immunotherapy for resectable NSCLC, stimulating fresh perspectives and delving into its merits and challenges in clinical application.
.
Collapse
|
22
|
Reyes-Gibby CC, Caterino JM, Coyne CJ, Kyriacou DN, Qdaisat A, McQuade J, Owen DH, Bischof JJ, Shete S, Yeung SCJ. Immune-related adverse event in the emergency department: methodology of the immune-related emergency disposition index (IrEDi). EMERGENCY CANCER CARE 2024; 3:1. [PMID: 38725994 PMCID: PMC11081141 DOI: 10.1186/s44201-023-00023-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/13/2023] [Indexed: 05/12/2024]
Abstract
For many cancer patients, immune checkpoint inhibitors (ICIs) can be life-saving. However, the immune-related adverse events (irAEs) from ICIs can be debilitating and can quickly become severe or even be fatal. Often, irAEs will precipitate visits to the emergency department (ED). Therefore, early recognition and the decision to admit, observe, or discharge these patients from the ED can be key to a cancer patient's morbidity and mortality. ED clinicians typically make their decision for disposition (admit, observe, or discharge) within 2-6 h from their patient's ED presentation. However, irAEs are particularly challenging in the ED because of atypical presentations, the absence of classic symptoms, the delayed availability of diagnostic tests during the ED encounter, and the fast pace in the ED setting. At present, there is no single sufficiently large ED data source with clinical, biological, laboratory, and imaging data that will allow for the development of a tool that will guide early recognition and appropriate ED disposition of patients with potential irAEs. We describe an ongoing federally funded project that aims to develop an immune-related emergency disposition index (IrEDi). The project capitalizes on a multi-site collaboration among 4 members of the Comprehensive Oncologic Emergency Research Network (CONCERN): MD Anderson Cancer Center, Ohio State University, Northwestern University, and University of California San Diego. If the aims are achieved, the IrEDi will be the first risk stratification tool derived from a large racial/ethnically and geographically diverse population of cancer patients. The future goal is to validate irEDi in general EDs to improve emergency care of cancer patients on ICIs.
Collapse
Affiliation(s)
- Cielito C. Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey M. Caterino
- Departments of Emergency Medicine and Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Christopher J. Coyne
- Department of Emergency Medicine, University of California San Diego, San Diego, CA, USA
| | - Demetrios N. Kyriacou
- Department of Emergency Medicine and Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Aiham Qdaisat
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dwight H. Owen
- Departments of Emergency Medicine and Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jason J. Bischof
- Departments of Emergency Medicine and Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sanjay Shete
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
23
|
Kovacsovics-Bankowski M, Sweere JM, Healy CP, Sigal N, Cheng LC, Chronister WD, Evans SA, Marsiglio J, Gibson B, Swami U, Erickson-Wayman A, McPherson JP, Derose YS, Eliason AL, Medina CO, Srinivasan R, Spitzer MH, Nguyen N, Hyngstrom J, Hu-Lieskovan S. Lower frequencies of circulating suppressive regulatory T cells and higher frequencies of CD4 + naïve T cells at baseline are associated with severe immune-related adverse events in immune checkpoint inhibitor-treated melanoma. J Immunother Cancer 2024; 12:e008056. [PMID: 38233101 PMCID: PMC10806651 DOI: 10.1136/jitc-2023-008056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Immune-related adverse events (irAEs) are major barriers of clinical management and further development of immune checkpoint inhibitors (ICIs) for cancer therapy. Therefore, biomarkers associated with the onset of severe irAEs are needed. In this study, we aimed to identify immune features detectable in peripheral blood and associated with the development of severe irAEs that required clinical intervention. METHODS We used a 43-marker mass cytometry panel to characterize peripheral blood mononuclear cells from 28 unique patients with melanoma across 29 lines of ICI therapy before treatment (baseline), before the onset of irAEs (pre-irAE) and at the peak of irAEs (irAE-max). In the 29 lines of ICI therapy, 18 resulted in severe irAEs and 11 did not. RESULTS Unsupervised and gated population analysis showed that patients with severe irAEs had a higher frequency of CD4+ naïve T cells and lower frequency of CD16+ natural killer (NK) cells at all time points. Gated population analysis additionally showed that patients with severe irAEs had fewer T cell immunoreceptor with Ig and ITIM domain (TIGIT+) regulatory T cells at baseline and more activated CD38+ CD4+ central memory T cells (TCM) and CD39+ and Human Leukocyte Antigen-DR Isotype (HLA-DR)+ CD8+ TCM at peak of irAEs. The differentiating immune features at baseline were predominantly seen in patients with gastrointestinal and cutaneous irAEs and type 1 diabetes. Higher frequencies of CD4+ naïve T cells and lower frequencies of CD16+ NK cells were also associated with clinical benefit to ICI therapy. CONCLUSIONS This study demonstrates that high-dimensional immune profiling can reveal novel blood-based immune signatures associated with risk and mechanism of severe irAEs. Development of severe irAEs in melanoma could be the result of reduced immune inhibitory capacity pre-ICI treatment, resulting in more activated TCM cells after treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John Marsiglio
- The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Berit Gibson
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Umang Swami
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alyssa Erickson-Wayman
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jordan P McPherson
- Department of Pharmacy, Huntsman Cancer Institute Cancer Hospital, Salt Lake City, Utah, USA
| | - Yoko S Derose
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | | | | | | | - Matthew H Spitzer
- Teiko.bio Inc, Salt Lake City, Utah, USA
- Department of Otolaryngology-Head and Neck Cancer, University of California San Francisco, San Francisco, California, USA
| | | | - John Hyngstrom
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Siwen Hu-Lieskovan
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
24
|
Möhn N, Renovanz M, Hagin D, Skripuletz T. Editorial: Autoimmune complications of modern cancer therapies. Front Immunol 2024; 14:1357825. [PMID: 38250085 PMCID: PMC10796640 DOI: 10.3389/fimmu.2023.1357825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Affiliation(s)
- Nora Möhn
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Immune Cooperative Oncology Group, Comprehensive Cancer Center Hannover (ICOG-CCCH), Hannover, Germany
| | - Mirjam Renovanz
- Department of Neurology & Interdisciplinary Neuro-Oncology, University Hospital Tuebingen, Hertie Institute for Clinical Brain Research, Tuebingen, Germany
- Department of Neurosurgery, University Hospital Tuebingen, Tuebingen, Germany
| | - David Hagin
- Department of Immunology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Immune Cooperative Oncology Group, Comprehensive Cancer Center Hannover (ICOG-CCCH), Hannover, Germany
| |
Collapse
|
25
|
Pernot S, Tomé M, Galeano-Otero I, Evrard S, Badiola I, Delom F, Fessart D, Smani T, Siegfried G, Villoutreix BO, Khatib AM. Sulconazole inhibits PD-1 expression in immune cells and cancer cells malignant phenotype through NF-κB and calcium activity repression. Front Immunol 2024; 14:1278630. [PMID: 38250065 PMCID: PMC10796450 DOI: 10.3389/fimmu.2023.1278630] [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: 08/16/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
The overexpression of the immunoinhibitory receptor programmed death-1 (PD1) on T-cells is involved in immune evasion in cancer. The use of anti-PD-1/PDL-1 strategy has deeply changed the therapies of cancers and patient survival. However, their efficacy diverges greatly along with tumor type and patient populations. Thereby, novel treatments are needed to interfere with the anti-tumoral immune responses and propose an adjunct therapy. In the current study, we found that the antifungal drug Sulconazole (SCZ) inhibits PD-1 expression on activated PBMCs and T cells at the RNA and protein levels. SCZ repressed NF-κB and calcium signaling, both, involved in the induction of PD-1. Further analysis revealed cancer cells treatment with SCZ inhibited their proliferation, and migration and ability to mediate tumor growth in zebrafish embryos. SCZ found also to inhibit calcium mobilization in cancer cells. These results suggest the SCZ therapeutic potential used alone or as adjunct strategy to prevent T-cell exhaustion and promotes cancer cell malignant phenotype repression in order to improve tumor eradication.
Collapse
Affiliation(s)
- Simon Pernot
- Reprograming tumor activitY and associaTed MicroenvironmEnt (Rytme), Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, Université of Bordeaux, Pessac, France
- Institut Bergonié, Bordeaux, France
| | - Mercedes Tomé
- Reprograming tumor activitY and associaTed MicroenvironmEnt (Rytme), Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, Université of Bordeaux, Pessac, France
| | - Isabel Galeano-Otero
- Reprograming tumor activitY and associaTed MicroenvironmEnt (Rytme), Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, Université of Bordeaux, Pessac, France
| | - Serge Evrard
- Reprograming tumor activitY and associaTed MicroenvironmEnt (Rytme), Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, Université of Bordeaux, Pessac, France
- Institut Bergonié, Bordeaux, France
| | - Iker Badiola
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Frederic Delom
- Reprograming tumor activitY and associaTed MicroenvironmEnt (Rytme), Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, Université of Bordeaux, Pessac, France
- Institut Bergonié, Bordeaux, France
| | - Delphine Fessart
- Reprograming tumor activitY and associaTed MicroenvironmEnt (Rytme), Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, Université of Bordeaux, Pessac, France
- Institut Bergonié, Bordeaux, France
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
| | - Geraldine Siegfried
- Reprograming tumor activitY and associaTed MicroenvironmEnt (Rytme), Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, Université of Bordeaux, Pessac, France
- Institut Bergonié, Bordeaux, France
| | - Bruno O. Villoutreix
- Integrative Computational Pharmacology and Data Mining, INSERM UMR 1141, Rob-ert-Debré Hospital, Paris, France
| | - Abdel-Majid Khatib
- Reprograming tumor activitY and associaTed MicroenvironmEnt (Rytme), Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, Université of Bordeaux, Pessac, France
- Institut Bergonié, Bordeaux, France
| |
Collapse
|
26
|
Li Y, Shu Y, Yu K, Ni R, Chu L. Analysis and Regulatory Mechanisms of Platelet-Related Genes in Patients with Ischemic Stroke. Cell Mol Neurobiol 2024; 44:15. [PMID: 38175286 PMCID: PMC11397819 DOI: 10.1007/s10571-023-01433-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
It was found that ischemic stroke (IS) was associated with abnormal platelet activity and thrombosis. However, the potential significance of platelet-related genes (PRGs) in IS still needs to be more thorough. This study extracted IS-related transcriptome datasets from the Gene Expression Omnibus (GEO) database. The target genes were obtained by intersecting the differentially expressed genes (DEGs), the module genes related to IS, and PRGs, where the key genes of IS were screened by two machine learning algorithms. The key genes-based diagnostic model was constructed. Gene set enrichment analysis (GSEA) and the immune microenvironment analyses were analyzed targeting key genes in IS. The co-expression, TF-mRNA, and competitive endogenous RNAs (ceRNA) regulatory networks were constructed to reveal the potential regulation of key genes. Potential drugs targeting key genes were predicted as well. Totals of eight target genes were obtained and were associated with immune-related functions. Four platelet-related key genes were acquired, which were related to immunity and energy metabolism. The abnormal expressions of DOCK8, GIMAP5, ICOS were determined by the quantitative real-time polymerase chain reaction (qRT-PCR), and the significant correlations among these key genes were identified. Notably, hsa-miR-17-3p, hsa-miR-3158-3p, hsa-miR-423-3p, and hsa-miR-193a-8p could regulate all key genes at the same time. In addition, Caffeine, Carboplatin, and Vopratelimab were the targeted drugs of these key genes. This study identified four platelet-related key genes of IS, which might help to deepen the understanding of the role of platelet-related genes in the molecular mechanism of IS.
Collapse
Affiliation(s)
- Yuan Li
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yuanlu Shu
- Department of Neurology, Guizhou Medical University, Guiyang, 550004, China
| | - Kun Yu
- Department of Neurology, Guizhou Medical University, Guiyang, 550004, China
| | - Ruihan Ni
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Lan Chu
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
- Department of Neurology, Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
27
|
Furuno T, Sogawa R, Hashimoto T, Matsuo S, Shirahama W, Kamura T, Hosoya K, Senjyu Y, Yamashita Y, Inoue T, Yamauchi M, Katsuya H, Noguchi M, Sueoka-Aragane N, Shimanoe C. Association between the Prognostic Nutritional Index and the Occurrence of Immune-Related Adverse Events. Biol Pharm Bull 2024; 47:361-365. [PMID: 38311396 DOI: 10.1248/bpb.b23-00760] [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: 02/10/2024]
Abstract
Immune-related adverse events (irAEs) affect all organs and are associated with various symptoms. The identification of biomarkers that can predict irAEs may be particularly clinically useful. This study aimed to investigate whether the prognostic nutritional index (PNI) before the initiation of immune checkpoint inhibitor (ICI) treatment can predict the occurrence of irAEs. We conducted a survey of 111 patients with cancer who were receiving ICI fixed-dose monotherapy at Saga University Hospital from the time each ICI became available until January 2020. We compared the PNI between the patients with and without irAE expression, established a cutoff value for PNI associated with the development of irAEs, and investigated the incidence of irAEs and progression-free survival (PFS) in groups divided by the cutoff value. Patients with irAEs had significantly higher PNI than did those without, and there was a significant association between PNI and irAEs after adjusting for potential factors (odds ratio, 1.12; 95% confidence interval, 1.03-1.21). In addition, PNI ≥44.2 was associated with a significantly higher incidence of irAEs (75.0% vs. 35.2%, p = 0.0001) and significantly longer PFS than PNI <44.2 (p = 0.025). In conclusion, pretreatment PNI may be associated with the risk of developing irAEs in patients with advanced recurrent solid tumors. When the PNI is ≥44.2, patient management is important for avoiding serious AEs because while the treatment may be effective, the occurrence of irAEs is a concern.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yoko Senjyu
- Department of Pharmacy, Saga University Hospital
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Rivera J, Digklia A, Christou AS, Anibal J, Vallis KA, Wood BJ, Stride E. A Review of Ultrasound-Mediated Checkpoint Inhibitor Immunotherapy. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1-7. [PMID: 37798210 DOI: 10.1016/j.ultrasmedbio.2023.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/11/2023] [Accepted: 08/26/2023] [Indexed: 10/07/2023]
Abstract
Over the past decade, immunotherapy has emerged as a major modality in cancer medicine. However, despite its unprecedented success, immunotherapy currently benefits only a subgroup of patients, may induce responses of limited duration and is associated with potentially treatment-limiting side effects. In addition, responses to immunotherapeutics are sometimes diminished by the emergence of a complex array of resistance mechanisms. The efficacy of immunotherapy depends on dynamic interactions between tumour cells and the immune landscape in the tumour microenvironment. Ultrasound, especially in conjunction with cavitation-promoting agents such as microbubbles, can assist in the uptake and/or local release of immunotherapeutic agents at specific target sites, thereby increasing treatment efficacy and reducing systemic toxicity. There is also increasing evidence that ultrasound and/or cavitation may themselves directly stimulate a beneficial immune response. In this review, we summarize the latest developments in the use of ultrasound and cavitation agents to promote checkpoint inhibitor immunotherapy.
Collapse
Affiliation(s)
- Jocelyne Rivera
- Center for Interventional Oncology, Interventional Radiology, National Institutes of Health Clinical Center, National Cancer Institute, Bethesda, MD, USA; Botnar Research Centre, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Antonia Digklia
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Anna S Christou
- Center for Interventional Oncology, Interventional Radiology, National Institutes of Health Clinical Center, National Cancer Institute, Bethesda, MD, USA
| | - James Anibal
- Center for Interventional Oncology, Interventional Radiology, National Institutes of Health Clinical Center, National Cancer Institute, Bethesda, MD, USA; Computational Health Informatics Lab, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | | | - Bradford J Wood
- Center for Interventional Oncology, Interventional Radiology, National Institutes of Health Clinical Center, National Cancer Institute, Bethesda, MD, USA
| | - Eleanor Stride
- Botnar Research Centre, Institute of Biomedical Engineering, University of Oxford, Oxford, UK.
| |
Collapse
|
29
|
Ma W, Wei S, Long S, Tian EC, McLaughlin B, Jaimes M, Montoya DJ, Viswanath VR, Chien J, Zhang Q, Van Dyke JE, Chen S, Li T. Dynamic evaluation of blood immune cells predictive of response to immune checkpoint inhibitors in NSCLC by multicolor spectrum flow cytometry. Front Immunol 2023; 14:1206631. [PMID: 37638022 PMCID: PMC10449448 DOI: 10.3389/fimmu.2023.1206631] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/20/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Immune checkpoint inhibitors (ICIs) only benefit a subset of cancer patients, underlining the need for predictive biomarkers for patient selection. Given the limitations of tumor tissue availability, flow cytometry of peripheral blood mononuclear cells (PBMCs) is considered a noninvasive method for immune monitoring. This study explores the use of spectrum flow cytometry, which allows a more comprehensive analysis of a greater number of markers using fewer immune cells, to identify potential blood immune biomarkers and monitor ICI treatment in non-small-cell lung cancer (NSCLC) patients. Methods PBMCs were collected from 14 non-small-cell lung cancer (NSCLC) patients before and after ICI treatment and 4 healthy human donors. Using spectrum flow cytometry, 24 immune cell markers were simultaneously monitored using only 1 million PBMCs. The results were also compared with those from clinical flow cytometry and bulk RNA sequencing analysis. Results Our findings showed that the measurement of CD4+ and CD8+ T cells by spectrum flow cytometry matched well with those by clinical flow cytometry (Pearson R ranging from 0.75 to 0.95) and bulk RNA sequencing analysis (R=0.80, P=1.3 x 10-4). A lower frequency of CD4+ central memory cells before treatment was associated with a longer median progression-free survival (PFS) [Not reached (NR) vs. 5 months; hazard ratio (HR)=8.1, 95% confidence interval (CI) 1.5-42, P=0.01]. A higher frequency of CD4-CD8- double-negative (DN) T cells was associated with a longer PFS (NR vs. 4.45 months; HR=11.1, 95% CI 2.2-55.0, P=0.003). ICIs significantly changed the frequency of cytotoxic CD8+PD1+ T cells, DN T cells, CD16+CD56dim and CD16+CD56- natural killer (NK) cells, and CD14+HLDRhigh and CD11c+HLADR + monocytes. Of these immune cell subtypes, an increase in the frequency of CD16+CD56dim NK cells and CD14+HLADRhigh monocytes after treatment compared to before treatment were associated with a longer PFS (NR vs. 5 months, HR=5.4, 95% CI 1.1-25.7, P=0.03; 7.8 vs. 3.8 months, HR=5.7, 95% CI 169 1.0-31.7, P=0.04), respectively. Conclusion Our preliminary findings suggest that the use of multicolor spectrum flow cytometry helps identify potential blood immune biomarkers for ICI treatment, which warrants further validation.
Collapse
Affiliation(s)
- Weijie Ma
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine, Dartmouth, NH, United States
| | - Sixi Wei
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Siqi Long
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Eddie C. Tian
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Bridget McLaughlin
- University of California Davis, Flow cytometry Shared Resource, Davis, CA, United States
| | | | - Dennis J. Montoya
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, United States
| | - Varun R. Viswanath
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Jeremy Chien
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, United States
| | - Qianjun Zhang
- Beckman Coulter Life Sciences, San Jose, CA, United States
| | - Jonathan E. Van Dyke
- University of California Davis, Flow cytometry Shared Resource, Davis, CA, United States
| | - Shuai Chen
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, Davis, CA, United States
| | - Tianhong Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, United States
| |
Collapse
|
30
|
Meybodi SM, Farasati Far B, Pourmolaei A, Baradarbarjastehbaf F, Safaei M, Mohammadkhani N, Samadani AA. Immune checkpoint inhibitors promising role in cancer therapy: clinical evidence and immune-related adverse events. Med Oncol 2023; 40:243. [PMID: 37453930 DOI: 10.1007/s12032-023-02114-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
The advent of immune checkpoint inhibitors (ICIs) has led to noteworthy progressions in the management of diverse cancer types, as evidenced by the pioneering "ipilimumab" medication authorized by US FDA in 2011. Importantly, ICIs agents have demonstrated encouraging potential in bringing about transformation across diverse forms of cancer by selectively targeting the immune checkpoint pathways that are exploited by cancerous cells for dodging the immune system, culminating in progressive and favorable health outcomes for patients. The primary mechanism of action (MOA) of ICIs involves blocking inhibitory immune checkpoints. There are three approved categories including Programmed Death (PD-1) inhibitors (cemiplimab, nivolumab, and pembrolizumab), Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) (Ipilimumab), and Programmed Death-Ligand 1 (PDL-1) (Avelumab). Although ICIs promisingly increase therapeutic response and cancer survival rates, using ICIs has demonstrated some limitations including autoimmune reactions and toxicities, requiring close monitoring. The present review endeavors to explicate the underlying principles of the MOA and pharmacokinetics of the approved ICIs in the realm of cancer induction, including an appraisal of their level of practice-based evidence.
Collapse
Affiliation(s)
| | - Bahareh Farasati Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, 1684613114, Iran.
| | - Ali Pourmolaei
- Babol Noshirvani University of Technology, Shariati Ave, Babol, Mazandaran, Iran
| | - Farid Baradarbarjastehbaf
- Faculty of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Pécs, Pécs, Hungary
| | - Maryam Safaei
- Department of Pharmacology, Faculty of Pharmacy, Eastern Mediterranean University, 99628, Famagusta, Turkey
| | - Niloufar Mohammadkhani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
31
|
Stephens MR, Asdourian MS, Jacoby TV, Shah N, Thompson LL, Otto T, Semenov YR, Reynolds KL, Sullivan RJ, Foreman RK, Chen ST. Tumor-infiltrating lymphocytes as a predictive biomarker of cutaneous immune-related adverse events after immune checkpoint blockade in patients with advanced melanoma. J Am Acad Dermatol 2023; 89:140-142. [PMID: 36806644 PMCID: PMC10796159 DOI: 10.1016/j.jaad.2023.01.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/28/2022] [Accepted: 01/13/2023] [Indexed: 02/17/2023]
Affiliation(s)
- Michael R Stephens
- Harvard Medical School, Boston, Massachusetts; Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Combined Dermatology Residency, Boston, Massachusetts
| | - Maria S Asdourian
- Harvard Medical School, Boston, Massachusetts; Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ted V Jacoby
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts; University of Hawaii at Manoa John A. Burns School of Medicine, Honolulu, Hawaii
| | - Nishi Shah
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts; Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Leah L Thompson
- Harvard Medical School, Boston, Massachusetts; Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| | - Tracey Otto
- Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Yevgeniy R Semenov
- Harvard Medical School, Boston, Massachusetts; Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| | - Kerry L Reynolds
- Harvard Medical School, Boston, Massachusetts; Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Ryan J Sullivan
- Harvard Medical School, Boston, Massachusetts; Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Ruth K Foreman
- Harvard Medical School, Boston, Massachusetts; Dermatopathology Service, Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Steven T Chen
- Harvard Medical School, Boston, Massachusetts; Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
32
|
Bhujel B, Oh SH, Kim CM, Yoon YJ, Kim YJ, Chung HS, Ye EA, Lee H, Kim JY. Mesenchymal Stem Cells and Exosomes: A Novel Therapeutic Approach for Corneal Diseases. Int J Mol Sci 2023; 24:10917. [PMID: 37446091 DOI: 10.3390/ijms241310917] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
The cornea, with its delicate structure, is vulnerable to damage from physical, chemical, and genetic factors. Corneal transplantation, including penetrating and lamellar keratoplasties, can restore the functions of the cornea in cases of severe damage. However, the process of corneal transplantation presents considerable obstacles, including a shortage of available donors, the risk of severe graft rejection, and potentially life-threatening complications. Over the past few decades, mesenchymal stem cell (MSC) therapy has become a novel alternative approach to corneal regeneration. Numerous studies have demonstrated the potential of MSCs to differentiate into different corneal cell types, such as keratocytes, epithelial cells, and endothelial cells. MSCs are considered a suitable candidate for corneal regeneration because of their promising therapeutic perspective and beneficial properties. MSCs compromise unique immunomodulation, anti-angiogenesis, and anti-inflammatory properties and secrete various growth factors, thus promoting corneal reconstruction. These effects in corneal engineering are mediated by MSCs differentiating into different lineages and paracrine action via exosomes. Early studies have proven the roles of MSC-derived exosomes in corneal regeneration by reducing inflammation, inhibiting neovascularization, and angiogenesis, and by promoting cell proliferation. This review highlights the contribution of MSCs and MSC-derived exosomes, their current usage status to overcome corneal disease, and their potential to restore different corneal layers as novel therapeutic agents. It also discusses feasible future possibilities, applications, challenges, and opportunities for future research in this field.
Collapse
Affiliation(s)
- Basanta Bhujel
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Se-Heon Oh
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Chang-Min Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Ye-Ji Yoon
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Young-Jae Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Ho-Seok Chung
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Eun-Ah Ye
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Hun Lee
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| | - Jae-Yong Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro, Songpa-Gu, Seoul 05505, Republic of Korea
| |
Collapse
|
33
|
Zhong J, Sun Z, Li S, Yang L, Cao Y, Bao J. Immune checkpoint blockade therapy for BRAF mutant metastatic colorectal cancer: the efficacy, new strategies, and potential biomarkers. Discov Oncol 2023; 14:94. [PMID: 37302081 PMCID: PMC10258190 DOI: 10.1007/s12672-023-00718-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023] Open
Abstract
BRAF mutant metastatic colorectal cancer has long been considered a tumor with a poor prognosis and a poor response to chemotherapy. Despite the efficacy of targeted therapy with multi-targeted blockade of the mitogen-activated protein kinase (MAPK) signaling pathway has brought a glimmer of hope to this group of patients, the need to improve treatment efficacy remains unmet, especially for the microsatellite stability/DNA proficient mismatch repair (MSS/pMMR) subtype. BRAF mutant colorectal cancer patients with high microsatellite instability/DNA deficient mismatch repair (MSI-H/dMMR) have high tumor mutation burden and abundant neoantigen, who are deemed as ones that could receive expected efficacy from immunotherapy. Generally, it is believed that MSS/pMMR colorectal cancer is an immunologically "cold" tumor that is insensitive to immunotherapy. However, targeted therapy combined with immune checkpoint blockade therapy seems to bring light to BRAF mutant colorectal cancer patients. In this review, we provide an overview of clinical efficacy and evolving new strategies concerning immune checkpoint blockade therapy for both MSI-H/dMMR and MSS/pMMR BRAF mutant metastatic colorectal cancer and discuss the potential biomarkers in the tumor immune microenvironment for predicting immunotherapeutic response in BRAF mutant colorectal cancer.
Collapse
Affiliation(s)
- Jie Zhong
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Zijian Sun
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Sheng Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Liu Yang
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Yuepeng Cao
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Jun Bao
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China.
| |
Collapse
|
34
|
Hosoda S, Suda G, Sho T, Ogawa K, Kimura M, Yang Z, Yoshida S, Kubo A, Tokuchi Y, Kitagataya T, Maehara O, Ohnishi S, Nakamura A, Yamada R, Ohara M, Kawagishi N, Natsuizaka M, Nakai M, Morikawa K, Furuya K, Baba M, Yamamoto Y, Suzuki K, Izumi T, Meguro T, Terashita K, Ito J, Miyagishima T, Sakamoto N. Low Baseline CXCL9 Predicts Early Progressive Disease in Unresectable HCC with Atezolizumab Plus Bevacizumab Treatment. Liver Cancer 2023; 12:156-170. [PMID: 37325489 PMCID: PMC10267515 DOI: 10.1159/000527759] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 10/13/2022] [Indexed: 07/30/2023] Open
Abstract
INTRODUCTION Atezolizumab plus bevacizumab treatment is highly effective in patients with unresectable hepatocellular carcinoma (HCC). However, progressive disease (PD) occurs in approximately 20% of HCC patients treated with atezolizumab plus bevacizumab, resulting in a poor prognosis. Thus, the prediction and early detection of HCC is crucial. METHODS Patients with unresectable HCC treated with atezolizumab plus bevacizumab and had baseline preserved serum (n = 68) were screened and classified according to their PD, 6 weeks after treatment initiation (early PD; n = 13). Of these, 4 patients each with and without early PD were selected for cytokine array and genetic analyses. The identified factors were validated in the validated cohort (n = 60) and evaluated in patients treated with lenvatinib. RESULTS No significant differences were observed in the genetic alterations in circulating tumor DNA. Cytokine array data revealed that baseline MIG (CXCL9), ENA-78, and RANTES differed substantially between patients with and without early PD. Subsequent analysis in the validation cohort revealed that baseline CXCL9 was significantly lower in patients with early PD than that in patients without early PD, and the best cut-off value of serum CXCL9 to predict early PD was 333 pg/mL (sensitivity: 0.600, specificity: 0.923, AUC = 0.75). In patients with lower serum CXCL9 (<333 pg/mL), 35.3% (12/34) experienced early PD with atezolizumab plus bevacizumab, while progression-free survival (PFS) was significantly shorter relative to that in patients without (median PFS, 126 days vs. 227 days; HR: 2.41, 95% CI: 1.22-4.80, p = 0.0084). While patients with objective response to lenvatinib had significantly lower CXCL9 levels compared with those of patients without. CONCLUSION Baseline low serum CXCL9 (<333 pg/mL) levels may predict early PD in patients with unresectable HCC treated with atezolizumab plus bevacizumab.
Collapse
Affiliation(s)
- Shunichi Hosoda
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Goki Suda
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takuya Sho
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Koji Ogawa
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Megumi Kimura
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Zijian Yang
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sonoe Yoshida
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akinori Kubo
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshimasa Tokuchi
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takashi Kitagataya
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Osamu Maehara
- Laboratory of Molecular and Cellular Medicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Shunsuke Ohnishi
- Laboratory of Molecular and Cellular Medicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Akihisa Nakamura
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ren Yamada
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masatsugu Ohara
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoki Kawagishi
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mitsuteru Natsuizaka
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masato Nakai
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kenichi Morikawa
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ken Furuya
- Department of Gastroenterology and Hepatology, Japan Community Health Care Organization Hokkaido Hospital, Hokkaido, Japan
| | - Masaru Baba
- Department of Gastroenterology and Hepatology, Japan Community Health Care Organization Hokkaido Hospital, Hokkaido, Japan
| | | | - Kazuharu Suzuki
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Hakodate City Hospital, Hokkaido, Japan
| | | | | | - Katsumi Terashita
- Japan Community Health Care Organization Sapporo Hokushin Hospital, Hokkaido, Japan
| | - Jun Ito
- The Hokkaido Medical Center, Hokkaido, Japan
| | | | - Naoya Sakamoto
- Departments of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
35
|
Lechner MG, Zhou Z, Hoang AT, Huang N, Ortega J, Scott LN, Chen HC, Patel AY, Yakhshi-Tafti R, Kim K, Hugo W, Famini P, Drakaki A, Ribas A, Angell TE, Su MA. Clonally expanded, thyrotoxic effector CD8 + T cells driven by IL-21 contribute to checkpoint inhibitor thyroiditis. Sci Transl Med 2023; 15:eadg0675. [PMID: 37196065 PMCID: PMC10227862 DOI: 10.1126/scitranslmed.adg0675] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/19/2023] [Indexed: 05/19/2023]
Abstract
Autoimmune toxicity occurs in up to 60% of patients treated with immune checkpoint inhibitor (ICI) therapy for cancer and represents an increasing clinical challenge for expanding the use of these treatments. To date, human immunopathogenic studies of immune-related adverse events (IRAEs) have relied on sampling of circulating peripheral blood cells rather than affected tissues. Here, we directly obtained thyroid specimens from individuals with ICI-thyroiditis, one of the most common IRAEs, and compared immune infiltrates with those from individuals with spontaneous autoimmune Hashimoto's thyroiditis (HT) or no thyroid disease. Single-cell RNA sequencing revealed a dominant, clonally expanded population of thyroid-infiltrating cytotoxic CXCR6+ CD8+ T cells (effector CD8+ T cells) present in ICI-thyroiditis but not HT or healthy controls. Furthermore, we identified a crucial role for interleukin-21 (IL-21), a cytokine secreted by intrathyroidal T follicular (TFH) and T peripheral helper (TPH) cells, as a driver of these thyrotoxic effector CD8+ T cells. In the presence of IL-21, human CD8+ T cells acquired the activated effector phenotype with up-regulation of the cytotoxic molecules interferon-γ (IFN-γ) and granzyme B, increased expression of the chemokine receptor CXCR6, and thyrotoxic capacity. We validated these findings in vivo using a mouse model of IRAEs and further demonstrated that genetic deletion of IL-21 signaling protected ICI-treated mice from thyroid immune infiltration. Together, these studies reveal mechanisms and candidate therapeutic targets for individuals who develop IRAEs.
Collapse
Affiliation(s)
- Melissa G. Lechner
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Zikang Zhou
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Aline T. Hoang
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
- Drexel Medical School; Philadelphia, PA 19129
| | - Nicole Huang
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Jessica Ortega
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Lauren N. Scott
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Ho-Chung Chen
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Anushi Y. Patel
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Rana Yakhshi-Tafti
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
- Rosalind Franklin Medical School; Chicago, IL 60064
| | - Kristy Kim
- UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Willy Hugo
- Division of Dermatology, Department of Medicine, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Pouyan Famini
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Alexandra Drakaki
- Division of Hematology and Oncology, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Antoni Ribas
- Division of Hematology and Oncology, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Trevor E. Angell
- Division of Endocrinology and Diabetes, USC Keck School of Medicine; Los Angeles, CA 90033
| | - Maureen A. Su
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
- Division of Pediatric Endocrinology, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| |
Collapse
|
36
|
Jiang M, Fiering S, Shao Q. Combining energy-based focal ablation and immune checkpoint inhibitors: preclinical research and clinical trials. Front Oncol 2023; 13:1153066. [PMID: 37251920 PMCID: PMC10211342 DOI: 10.3389/fonc.2023.1153066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/12/2023] [Indexed: 05/31/2023] Open
Abstract
Energy-based focal therapy (FT) uses targeted, minimally invasive procedures to destroy tumors while preserving normal tissue and function. There is strong emerging interest in understanding how systemic immunity against the tumor can occur with cancer immunotherapy, most notably immune checkpoint inhibitors (ICI). The motivation for combining FT and ICI in cancer management relies on the synergy between the two different therapies: FT complements ICI by reducing tumor burden, increasing objective response rate, and reducing side effects of ICI; ICI supplements FT by reducing local recurrence, controlling distal metastases, and providing long-term protection. This combinatorial strategy has shown promising results in preclinical study (since 2004) and the clinical trials (since 2011). Understanding the synergy calls for understanding the physics and biology behind the two different therapies with distinctive mechanisms of action. In this review, we introduce different types of energy-based FT by covering the biophysics of tissue-energy interaction and present the immunomodulatory properties of FT. We discuss the basis of cancer immunotherapy with the emphasis on ICI. We examine the approaches researchers have been using and the results from both preclinical models and clinical trials from our exhaustive literature research. Finally, the challenges of the combinatory strategy and opportunities of future research is discussed extensively.
Collapse
Affiliation(s)
- Minhan Jiang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Dartmouth Cancer Center, Dartmouth Geisel School of Medicine and Dartmouth Health, Lebanon, NH, United States
| | - Qi Shao
- Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
37
|
Mohanty SK, Mishra SK, Amin MB, Agaimy A, Fuchs F. Role of Surgical Pathologist for the Detection of Immuno-oncologic Predictive Factors in Non-small Cell Lung Cancers. Adv Anat Pathol 2023; 30:174-194. [PMID: 37037418 DOI: 10.1097/pap.0000000000000395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Until very recently, surgery, chemotherapy, and radiation therapy have been the mainstay of treatment in non-small cell carcinomas (NSCLCs). However, recent advances in molecular immunology have unveiled some of the complexity of the mechanisms regulating cellular immune responses and led to the successful targeting of immune checkpoints in attempts to enhance antitumor T-cell responses. Immune checkpoint molecules such as cytotoxic T-lymphocyte associated protein-4, programmed cell death protein-1, and programmed death ligand (PD-L) 1 have been shown to play central roles in evading cancer immunity. Thus, these molecules have been targeted by inhibitors for the management of cancers forming the basis of immunotherapy. Advanced NSCLC has been the paradigm for the benefits of immunotherapy in any cancer. Treatment decisions are made based on the expression of PD-L1 on the tumor cells and the presence or absence of driver mutations. Patients with high PD-L1 expression (≥50%) and no driver mutations are treated with single-agent immunotherapy whereas, for all other patients with a lower level of PD-L1 expression, a combination of chemotherapy and immunotherapy is preferred. Thus, PD-L1 blockers are the only immunotherapeutic agents approved in advanced NSCLC without any oncogenic driver mutations. PD-L1 immunohistochemistry, however, may not be the best biomarker in view of its dynamic nature in time and space, and the benefits may be seen regardless of PD -L1 expression. Each immunotherapy molecule is prescribed based on the levels of PD-L1 expression as assessed by a Food and Drug Administration-approved companion diagnostic assay. Other biomarkers that have been studied include tumor mutational burden, the T-effector signature, tumor-infiltrating lymphocytes, radiomic assays, inflammation index, presence or absence of immune-related adverse events and specific driver mutations, and gut as well as local microbiome. At the current time, none of these biomarkers are routinely used in the clinical decision-making process for immunotherapy in NSCLC. However, in individual cases, they can be useful adjuncts to conventional therapy. This review describes our current understanding of the role of biomarkers as predictors of response to immune checkpoint molecules. To begin with a brief on cancer immunology in general and in NSCLC, in particular, is discussed. In the end, recent advancements in laboratory techniques for refining biomarker assays are described.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India and CORE Diagnostics, Gurgaon, HR
| | - Sourav K Mishra
- Department of Medical Oncology, All India Institute of Medical Sciences, DL, India
| | - Mahul B Amin
- Departments of Pathology and Laboratory Medicine and Urology, University of Tennessee Health Science Center, Memphis, TN
| | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Fuchs
- Department of Internal Medicine-1, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen University Hospital and Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Immune check point inhibitors (ICIs) are a unique class of cancer treatments that harness the body's innate antitumor response. Although these medications have transformed oncology care, they also lead to generalized immune activation that can result in toxicities across a spectrum of organ systems called immune-related adverse events. This article reviews the most common rheumatologic immune-related adverse events and their management. RECENT FINDINGS Inflammatory arthritis, polymyalgia rheumatic, sicca symptoms, systemic sclerosis, myositis, and vasculitis have all been reported as ICI adverse events. Treatment includes nonsteroidal anti-inflammatory drugs, glucocorticoids, traditional DMARDs, and biologics. SUMMARY Rheumatologists have an important role in the management of patients with rheumatologic immune-related adverse events. Working with our oncology colleagues, we can help manage rheumatologic immune-related adverse events while optimally preserving ICI's antitumor effects.
Collapse
Affiliation(s)
- Melissa Defoe
- Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
39
|
Mehra T, Dongre K, Boesing M, Frei P, Suenderhauf C, Zippelius A, Leuppi JD, Wicki A, Leuppi-Taegtmeyer AB. Pre-treatment comorbidities, C-reactive protein and eosinophil count, and immune-related adverse events as predictors of survival with checkpoint inhibition for multiple tumour entities. Cancer Med 2023. [PMID: 37084178 DOI: 10.1002/cam4.5919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND The development of immune-related adverse events (irAEs) may be associated with clinical efficacy of checkpoint inhibitors (CPIs) in patients with cancer. We therefore investigated the effect of irAEs and pre-treatment parameters on outcome in a large, real-life patient cohort. METHODS We performed a single-centre, retrospective, observational study including patients who received CPIs from 2011 to 2018 and followed until 2021. The primary outcome was overall survival, and the secondary outcome was the development of irAEs. RESULTS In total, 229 patients with different tumour entities (41% non-small cell lung cancer [NSCLC], 29% melanoma) received a total of 282 CPI treatment courses (ipilimumab, nivolumab, pembrolizumab or atezolizumab). Thirty-four percent of patients developed irAEs (of these 17% had CTCAE Grade ≥3). Factors independently associated with mortality were pre-treatment CRP ≥10 mg/L (hazard ratio [HR] 2.064, p = 0.0003), comorbidity measured by Charlson comorbidity index (HR 1.149, p = 0.014) and irAEs (HR 0.644, p = 0.036) (age-adjusted, n = 216). Baseline eosinophil count ≤0.2 × 109 /L was a further independent predictor of mortality (age-, CRP-, CCI- and irAE-adjusted HR = 2.252, p = 0.002, n = 166). Anti-CTLA-4 use (p < 0.001), and pre-treatment CRP <10 mg/L were independently associated with irAE occurrence (p = 0.037). CONCLUSIONS We found an independent association between irAE occurrence and improved overall survival in a real-life cohort spanning multiple tumour entities and treatment regimens. Pre-treatment comorbidities, CRP and eosinophil count represent potential markers for predicting treatment response.
Collapse
Affiliation(s)
- Tarun Mehra
- Department of Oncology, Medical University Clinic, Kantonsspital Baselland, Liestal, Switzerland
- Department of Oncology & Hematology, University Hospital Zürich, University of Zurich, Zürich, Switzerland
| | - Kanchan Dongre
- Department of Clinical Pharmacology & Toxicology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Patient Safety, University Hospital Basel, Basel, Switzerland
| | - Maria Boesing
- Medical University Clinic, Kantonsspital Baselland, Liestal, Switzerland
| | - Patricia Frei
- Department of Clinical Pharmacology & Toxicology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Claudia Suenderhauf
- Department of Clinical Pharmacology & Toxicology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alfred Zippelius
- Department of Oncology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Joerg D Leuppi
- Medical University Clinic, Kantonsspital Baselland, Liestal, Switzerland
| | - Andreas Wicki
- Department of Oncology, Medical University Clinic, Kantonsspital Baselland, Liestal, Switzerland
- Department of Oncology & Hematology, University Hospital Zürich, University of Zurich, Zürich, Switzerland
| | - Anne B Leuppi-Taegtmeyer
- Department of Clinical Pharmacology & Toxicology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Patient Safety, University Hospital Basel, Basel, Switzerland
- Medical University Clinic, Kantonsspital Baselland, Liestal, Switzerland
| |
Collapse
|
40
|
Dutta R, Rathor A, Sharma HP, Pandey HC, Malik PS, Mohan A, Nambirajan A, Kumar R, Jain D. Mapping the immune landscape in small cell lung cancer by analysing expression of immuno-modulators in tissue biopsies and paired blood samples. Sci Rep 2023; 13:3739. [PMID: 36879122 PMCID: PMC9988882 DOI: 10.1038/s41598-023-30841-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Small cell lung carcinomas (SCLC) are aggressive tumors with high propensity to metastasize. Recent NCCN guidelines have incorporated immunotherapy in extensive stage SCLC. Limited benefit in few patients compounded by side effects of unwonted immune-checkpoint-inhibitor (ICPI) usage necessitates identification of potential biomarkers predicting response to ICPIs. Attempting this, we analysed expression of various immunoregulatory molecules in tissue biopsies and paired blood samples of SCLC patients. In 40 cases, immunohistochemistry for expression of immune inhibitory receptors CTLA-4, PD-L1 and IDO1 was performed. Matched blood samples were quantified for IFN-γ, IL-2, TNF-α and sCTLA-4 levels using immunoassay and additionally for IDO1 activity (Kynurenine/Tryptophan ratio) using LC-MS. Immunopositivity for PD-L1, IDO1 and CTLA-4 was identified in 9.3%, 6.2% and 71.8% cases, respectively. Concentration of serum IFN-γ (p-value < 0.001), TNF-α (p-value = 0.025) and s-CTLA4 (p-value = 0.08) were higher in SCLC patients while IL-2 was lower (p-value = 0.003) as compared to healthy controls. IDO1 activity was significantly elevated in SCLC cohort (p-value = 0.007). We proffer that SCLC patients show immune suppressive milieu in their peripheral circulation. Analysis of CTLA4 immunohistochemical expression along with s-CTLA4 levels appears prospective as biomarkers for predicting responsiveness to ICPIs. Additionally, evaluation of IDO1 appears cogent both as prognostic marker and potential therapeutic target as well.
Collapse
Affiliation(s)
- Rimlee Dutta
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Amber Rathor
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Hanuman Prasad Sharma
- Bioanalytics Facility, Centralized Core Research Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Hem Chandra Pandey
- Department of Transfusion Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Prabhat Singh Malik
- Department of Medical Oncology, Dr B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Anant Mohan
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Aruna Nambirajan
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Rajeev Kumar
- Delhi Cancer Registry, Dr B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
41
|
Ren B, Shen J, Qian Y, Zhou T. Sarcopenia as a Determinant of the Efficacy of Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer: A Meta-Analysis. Nutr Cancer 2023; 75:685-695. [PMID: 36533715 DOI: 10.1080/01635581.2022.2153879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE The impact of pre-immunotherapy sarcopenia in patients with non-small cell lung cancer (NSCLC) receiving immune checkpoint inhibitors (ICIs) is elusive. We performed a meta-analysis to investigate the association between sarcopenia and clinical outcomes of ICIs. METHODS PubMed, EMBASE, and the Cochrane Library were searched. RESULTS Thirteen clinical trials were selected. The 1,2-year overall survival rate was lower in the sarcopenia group (odds ratio (OR) = 2.44, 95% confidence interval (CI), 1.78-3.35, P < 0.00001; OR = 1.60, 95% CI, 1.08-2.37, P = 0.02), with I2 = 34%, P = 0.15, and I2 = 41%, P = 0.12. The 1,2-year progression-free survival (PFS) was the same (OR = 3.43, 95% CI, 1.86-6.33, P < 0.0001; OR = 2.06, 95% CI, 1.19-3.58, P < 0.0001), with I2 = 31%, P = 0.17 and I2=31%, P = 0.17. Sarcopenia reduced the overall response rate (OR = 2.22, 95% CI, 1.01-4.84, P = 0.02), with I2= 56%, P = 0.02, and disease control rate (OR = 3.15, 95% CI, 2.10-4.72, P < 0.0001) with I2 = 33%, P = 0.18. CONCLUSION Pre-immunotherapy sarcopenia was associated with poor clinical outcomes in patients with advanced NSCLC who received ICIs.
Collapse
Affiliation(s)
- Bixin Ren
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiucheng Shen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yajuan Qian
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tong Zhou
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
42
|
Godoy LA, Chen J, Ma W, Lally J, Toomey KA, Rajappa P, Sheridan R, Mahajan S, Stollenwerk N, Phan CT, Cheng D, Knebel RJ, Li T. Emerging precision neoadjuvant systemic therapy for patients with resectable non-small cell lung cancer: current status and perspectives. Biomark Res 2023; 11:7. [PMID: 36650586 PMCID: PMC9847175 DOI: 10.1186/s40364-022-00444-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/16/2022] [Indexed: 01/19/2023] Open
Abstract
Over the past decade, targeted therapy for oncogene-driven NSCLC and immune checkpoint inhibitors for non-oncogene-driven NSCLC, respectively, have greatly improved the survival and quality of life for patients with unresectable NSCLC. Increasingly, these biomarker-guided systemic therapies given before or after surgery have been used in patients with early-stage NSCLC. In March 2022, the US FDA granted the approval of neoadjuvant nivolumab and chemotherapy for patients with stage IB-IIIA NSCLC. Several phase II/III trials are evaluating the clinical efficacy of various neoadjuvant immune checkpoint inhibitor combinations for non-oncogene-driven NSCLC and neoadjuvant molecular targeted therapies for oncogene-driven NSCLC, respectively. However, clinical application of precision neoadjuvant treatment requires a paradigm shift in the biomarker testing and multidisciplinary collaboration at the diagnosis of early-stage NSCLC. In this comprehensive review, we summarize the current diagnosis and treatment landscape, recent advances, new challenges in biomarker testing and endpoint selections, practical considerations for a timely multidisciplinary collaboration at diagnosis, and perspectives in emerging neoadjuvant precision systemic therapy for patients with resectable, early-stage NSCLC. These biomarker-guided neoadjuvant therapies hold the promise to improve surgical and pathological outcomes, reduce systemic recurrences, guide postoperative therapy, and improve cure rates in patients with resectable NSCLC.
Collapse
Affiliation(s)
- Luis A Godoy
- Division of Thoracic Surgery, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Joy Chen
- Medical Student, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Weijie Ma
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jag Lally
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Kyra A Toomey
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Prabhu Rajappa
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Roya Sheridan
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Shirish Mahajan
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Nicholas Stollenwerk
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
- Medical Service, Pulmonology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Chinh T Phan
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
- Medical Service, Pulmonology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Danny Cheng
- Department of Radiology, Interventional Radiology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Robert J Knebel
- Department of Radiology, Interventional Radiology, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Tianhong Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA.
- Medical Service, Hematology and Oncology, Veterans Affairs Northern California Health Care System, Mather, CA, USA.
| |
Collapse
|
43
|
Wu YH, Chen RJ, Chiu HW, Yang LX, Wang YL, Chen YY, Yeh YL, Liao MY, Wang YJ. Nanoparticles augment the therapeutic window of RT and immunotherapy for treating cancers: pivotal role of autophagy. Theranostics 2023; 13:40-58. [PMID: 36593951 PMCID: PMC9800737 DOI: 10.7150/thno.77233] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022] Open
Abstract
Immunotherapies are now emerging as an efficient anticancer therapeutic strategy. Cancer immunotherapy utilizes the host's immune system to fight against cancer cells and has gained increasing interest due to its durable efficacy and low toxicity compared to traditional antitumor treatments, such as chemotherapy and radiotherapy (RT). Although the combination of RT and immunotherapy has drawn extensive attention in the clinical setting, the overall response rates are still low. Therefore, strategies for further improvement are urgently needed. Nanotechnology has been used in cancer immunotherapy and RT to target not only cancer cells but also the tumor microenvironment (TME), thereby helping to generate a long-term immune response. Nanomaterials can be an effective delivery system and a strong autophagy inducer, with the ability to elevate autophagy to very high levels. Interestingly, autophagy could play a critical role in optimal immune function, mediating cell-extrinsic homeostatic effects through the regulation of danger signaling in neoplastic cells under immunogenic chemotherapy and/or RT. In this review, we summarize the preclinical and clinical development of the combination of immunotherapy and RT in cancer therapy and highlight the latest progress in nanotechnology for augmenting the anticancer effects of immunotherapy and RT. The underlying mechanisms of nanomaterial-triggered autophagy in tumor cells and the TME are discussed in depth. Finally, we suggest the implications of these three strategies combined together to achieve the goal of maximizing the therapeutic advantages of cancer therapy and show recent advances in biomarkers for tumor response in the evaluation of those therapies.
Collapse
Affiliation(s)
- Yuan-Hua Wu
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Rong-Jane Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Hui-Wen Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City 234, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 110, Taiwan
| | - Li-Xing Yang
- Institute of Oral Medicine and Department of Stomatology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 701, Taiwan
| | - Yung-Li Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Ya-Ling Yeh
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Mei-Yi Liao
- Department of Applied Chemistry, National Pingtung University, Pingtung 900, Taiwan
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
44
|
Lee M, Jeong K, Park YR, Rhee Y. Increased risk of incident diabetes after therapy with immune checkpoint inhibitor compared with conventional chemotherapy: A longitudinal trajectory analysis using a tertiary care hospital database. Metabolism 2023; 138:155311. [PMID: 36122764 DOI: 10.1016/j.metabol.2022.155311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Immune checkpoint inhibitor (ICI) has been emerged as a promising cancer treatment. However, ICI use induces immune-related adverse events, including diabetes mellitus. We compared the risk of new-onset diabetes between patients receiving an ICI and those receiving conventional chemotherapy (CC). METHODS Using a tertiary care hospital database, we included cancer patients without a previous history of diabetes who were treated with either CC or an ICI. One-to-five nearest neighbor propensity matching was applied, and the risk of diabetes was estimated using a Cox proportional hazards model. Latent class growth modeling was performed with a trajectory approach to determine distinct clusters that followed similar glucose trajectory patterns over time. RESULTS Among 1326 subjects, 1105 received CC, and 221 received an ICI. The risk of new-onset diabetes was significantly higher in the ICI group than the CC group (adjusted hazard ratio 2.454, 95 % confidence interval 1.528-3.940; p < 0.001). The ICI group had a higher proportion of subjects in the trajectory cluster with an increasing glucose pattern than the CC group (10.4 % and 7.4 %, respectively). Within the ICI group, the subjects with an increasing glucose pattern were predominantly male and associated with enhanced lymphocytosis after ICI administration. CONCLUSIONS ICI therapy is associated with an increased risk of incident diabetes compared with CC. The glucose levels of patients treated with an ICI, especially males and those with prominent lymphocytosis after ICI treatment, need to be monitored regularly to detect ICI-associated diabetes as early as possible.
Collapse
Affiliation(s)
- Minyoung Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyeongseob Jeong
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Rang Park
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Yumie Rhee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
45
|
Ohuchi K, Amagai R, Kambayashi Y, Asano Y, Fujimura T. Serum CCL22 Increased in Advanced Melanoma Patients with Liver Metastases: Report of 5 Cases. Case Rep Oncol 2022; 15:1114-1120. [PMID: 36605221 PMCID: PMC9808253 DOI: 10.1159/000528328] [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: 08/12/2022] [Accepted: 11/11/2022] [Indexed: 12/28/2022] Open
Abstract
Advanced melanoma patients with liver metastases show a limited response to immunotherapy by the induction of regulatory T cells and depletion of effector cells, which leads to a poor prognosis. Tumor-associated macrophages (TAMs) induce apoptosis of activated antigen-specific CD8+ T cells in melanomas, leading to induction of tolerance to immune checkpoint inhibitors. In addition, TAMs produce various chemokines, and several serum pro-inflammatory chemokines measured at baseline are useful for the prediction of the efficacy of immunomodulatory drugs. In this study, serum levels of CCL22, CXCL5, and CXCL10 were evaluated by ELISA at baseline in 10 melanoma patients, 5 with liver metastases and 5 with lung metastases, treated with anti-PD1 Abs. Serum levels of CCL22, but not CXCL5 and CXCL10, were increased in patients with liver metastases compared to those with lung metastases or historical controls. The present data suggest that elevated serum CCL22 levels might be a biomarker for liver metastases in melanoma patients.
Collapse
Affiliation(s)
- Kentaro Ohuchi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryo Amagai
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshihide Asano
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
46
|
Deng Y, Li H, Fu J, Pu Y, Zhang Y, Chen S, Tong S, Liu H. A hypoxia risk score for prognosis prediction and tumor microenvironment in adrenocortical carcinoma. Front Genet 2022; 13:796681. [PMID: 36583015 PMCID: PMC9792869 DOI: 10.3389/fgene.2022.796681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Adrenocortical carcinoma (ACC) is a rare malignant endocrine tumor derived from the adrenal cortex. Because of its highly aggressive nature, the prognosis of patients with adrenocortical carcinoma is not impressive. Hypoxia exists in the vast majority of solid tumors and contributes to invasion, metastasis, and drug resistance. This study aimed to reveal the role of hypoxia in Adrenocortical carcinoma and develop a hypoxia risk score (HRS) for Adrenocortical carcinoma prognostic prediction. Methods: Hypoxia-related genes were obtained from the Molecular Signatures Database. The training cohorts of patients with adrenocortical carcinoma were downloaded from The Cancer Genome Atlas, while another three validation cohorts with comprehensive survival data were collected from the Gene Expression Omnibus. In addition, we constructed a hypoxia classifier using a random survival forest model. Moreover, we explored the relationship between the hypoxia risk score and immunophenotype in adrenocortical carcinoma to evaluate the efficacy of immune check inhibitors (ICI) therapy and prognosis of patients. Results: HRS and tumor stage were identified as independent prognostic factors. HRS was negatively correlated with immune cycle activity, immune cell infiltration, and the T cell inflammatory score. Therefore, we considered the low hypoxia risk score group as the inflammatory immunophenotype, whereas the high HRS group was a non-inflammatory immunophenotype. In addition, the HRS was negatively related to the expression of common immune checkpoint molecules such as PD-L1, CD200, CTLA-4, and TIGIT, suggesting that patients with a lower hypoxia risk score respond better to immunotherapy. Conclusion: We developed and validated a novel hypoxia risk score to predict the immunophenotype and response of patients with adrenocortical carcinoma to immune check inhibitors therapy. These findings not only provide fresh prognostic indicators for adrenocortical carcinoma but also offer several promising treatment targets for this disease.
Collapse
Affiliation(s)
- Yuanyuan Deng
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinglan Fu
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Pu
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Zhang
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Shijing Chen
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China
| | - Shiyu Tong
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Huixia Liu, ; Shiyu Tong,
| | - Huixia Liu
- Department of Geriatric Endocrine, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Huixia Liu, ; Shiyu Tong,
| |
Collapse
|
47
|
Yang M, Zhang Q, Ge Y, Tang M, Zhang X, Song M, Ruan G, Zhang X, Zhang K, Shi H. Glucose to lymphocyte ratio predicts prognoses in patients with colorectal cancer. Asia Pac J Clin Oncol 2022. [DOI: 10.1111/ajco.13904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 10/11/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Ming Yang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| | - Qi Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| | - Yizhong Ge
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou China
| | - Meng Tang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| | - Xi Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| | - Mengmeng Song
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| | - Guotian Ruan
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| | - Xiaowei Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| | - Kangping Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| | - Hanping Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition Beijing Shijitan Hospital Capital Medical University Beijing China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition Beijing China
- Key Laboratory of Cancer FSMP for State Market Regulation Beijing China
| |
Collapse
|
48
|
Minegishi S, Kinguchi S, Horita N, Namkoong H, Briasoulis A, Ishigami T, Tamura K, Nishiyama A, Yano Y. Immune Checkpoint Inhibitors Do Not Increase Short-Term Risk of Hypertension in Cancer Patients: a Systematic Literature Review and Meta-Analysis. Hypertension 2022; 79:2611-2621. [DOI: 10.1161/hypertensionaha.122.19865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
Immune checkpoint inhibitors (ICIs) are becoming widely used for novel cancer treatments. Immune-related adverse events, including cardiac toxicity, are frequently observed following immune checkpoint inhibitor (ICI) use. However, little is known regarding the association between ICIs initiation and hypertension in cancer patients.
Methods:
A systematic literature search was performed using PubMed, EMBASE, Cochrane Library, and Web of Science Core Collection. The risk of hypertension associated with ICI initiation in randomized controlled trials (RCTs) was evaluated. Hypertension was categorized according to the Common Terminology Criteria for Adverse Events. The odds ratios of grades I to V and grades III to V hypertension were calculated using a random-effects meta-analysis.
Results:
Thirty-two RCTs (n=19 810 cancer patients) were included. At a median follow-up of 36 months, the median overall survival was 15 months in the ICI group. ICI initiation was not significantly associated with hypertension (grades I–V: odds ratio, 1.12 [95% CI, 0.96–1.30]; grades III–V: odds ratio, 0.95 [95% CI, 0.78–1.16]). Additionally, no significant differences in hypertension risk were evident in ICI combination therapies with various drugs, including anti-VEGF (vascular endothelial growth factor) agents. In a subgroup analysis based on clinical setting (placebo RCT versus nonplacebo RCT), there were discrepancies between the results obtained with different methodologies, with patients in the nonplacebo RCTs having higher grades I–V hypertension (I
2
=88.6%,
P
for heterogeneity=0.003).
Conclusions:
ICI initiation was not associated with short-term risk of hypertension in cancer patients, and the association was similar regardless of concomitant treatment with other anticancer drugs.
Collapse
Affiliation(s)
- Shintaro Minegishi
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University Graduate School of Medicine, Japan (S.M., S.K., T.I., K.T.)
| | - Sho Kinguchi
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University Graduate School of Medicine, Japan (S.M., S.K., T.I., K.T.)
| | - Nobuyuki Horita
- Chemotherapy Center, Yokohama City University Hospital, Japan (N.H.)
| | - Ho Namkoong
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan (H.N.)
| | - Alexandros Briasoulis
- Division of Cardiovascular Diseases, Section of Heart Failure and Transplant, University of Iowa College of Medicine, Iowa City (A.B.)
| | - Tomoaki Ishigami
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University Graduate School of Medicine, Japan (S.M., S.K., T.I., K.T.)
| | - Kouichi Tamura
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University Graduate School of Medicine, Japan (S.M., S.K., T.I., K.T.)
| | - Akira Nishiyama
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan (A.N.)
| | - Yuichiro Yano
- NCD Epidemiology Research Center (NERC), Shiga University of Medical Science, Otsu, Japan (Y.Y.)
| | | |
Collapse
|
49
|
Landeira-Viñuela A, Arias-Hidalgo C, Juanes-Velasco P, Alcoceba M, Navarro-Bailón A, Pedreira CE, Lecrevisse Q, Díaz-Muñoz L, Sánchez-Santos JM, Hernández ÁP, García-Vaquero ML, Góngora R, De Las Rivas J, González M, Orfao A, Fuentes M. Unravelling soluble immune checkpoints in chronic lymphocytic leukemia: Physiological immunomodulators or immune dysfunction. Front Immunol 2022; 13:965905. [PMID: 36248816 PMCID: PMC9554405 DOI: 10.3389/fimmu.2022.965905] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a lymphoid neoplasm characterized by the accumulation of mature B cells. The diagnosis is established by the detection of monoclonal B lymphocytes in peripheral blood, even in early stages [monoclonal B-cell lymphocytosis (MBLhi)], and its clinical course is highly heterogeneous. In fact, there are well-characterized multiple prognostic factors that are also related to the observed genetic heterogenicity, such as immunoglobulin heavy chain variable region (IGHV) mutational status, del17p, and TP53 mutations, among others. Moreover, a dysregulation of the immune system (innate and adaptive immunity) has been observed in CLL patients, with strong impact on immune surveillance and consequently on the onset, evolution, and therapy response. In addition, the tumor microenvironment is highly complex and heterogeneous (i.e., matrix, fibroblast, endothelial cells, and immune cells), playing a critical role in the evolution of CLL. In this study, a quantitative profile of 103 proteins (cytokines, chemokines, growth/regulatory factors, immune checkpoints, and soluble receptors) in 67 serum samples (57 CLL and 10 MBLhi) has been systematically evaluated. Also, differential profiles of soluble immune factors that discriminate between MBLhi and CLL (sCD47, sCD27, sTIMD-4, sIL-2R, and sULBP-1), disease progression (sCD48, sCD27, sArginase-1, sLAG-3, IL-4, and sIL-2R), or among profiles correlated with other prognostic factors, such as IGHV mutational status (CXCL11/I-TAC, CXCL10/IP-10, sHEVM, and sLAG-3), were deciphered. These results pave the way to explore the role of soluble immune checkpoints as a promising source of biomarkers in CLL, to provide novel insights into the immune suppression process and/or dysfunction, mostly on T cells, in combination with cellular balance disruption and microenvironment polarization leading to tumor escape.
Collapse
Affiliation(s)
- Alicia Landeira-Viñuela
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Carlota Arias-Hidalgo
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Pablo Juanes-Velasco
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Alcoceba
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Almudena Navarro-Bailón
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Carlos Eduardo Pedreira
- Systems and Computing Department Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia-Programa de Engenharia de Sistemas e Computação (COPPE-PESC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Quentin Lecrevisse
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Laura Díaz-Muñoz
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | | - Ángela-Patricia Hernández
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Department of Pharmaceutical Sciences, Organic Chemistry Section, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| | - Marina L. García-Vaquero
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Rafael Góngora
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center Instituto Universitario de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca (CIC-IBMCC, CSIC/USAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Salamanca, Spain
| | - Marcos González
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Manuel Fuentes
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Proteomics Unit, Cancer Research Centre-IBMCC, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca-Consejo Superior de Investigaciones Científicas (CSIC), Salamanca, Spain
- *Correspondence: Manuel Fuentes,
| |
Collapse
|
50
|
Lechner MG, Cheng MI, Patel AY, Hoang AT, Yakobian N, Astourian M, Pioso MS, Rodriguez ED, McCarthy EC, Hugo W, Angell TE, Drakaki A, Ribas A, Su MA. Inhibition of IL-17A Protects against Thyroid Immune-Related Adverse Events while Preserving Checkpoint Inhibitor Antitumor Efficacy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:696-709. [PMID: 35817515 PMCID: PMC9378719 DOI: 10.4049/jimmunol.2200244] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/20/2022] [Indexed: 11/19/2022]
Abstract
Immune checkpoint inhibitor (ICI) immunotherapy leverages the body's own immune system to attack cancer cells but leads to unwanted autoimmune side effects in up to 60% of patients. Such immune-related adverse events (IrAEs) may lead to treatment interruption, permanent organ dysfunction, hospitalization, and premature death. Thyroiditis is one of the most common IrAEs, but the cause of thyroid IrAEs remains unknown. In this study, we use a new, physiologically relevant mouse model of ICI-associated autoimmunity to identify a key role for type 3 immune cells in the development of thyroid IrAEs. Multiple lineages of IL-17A-producing T cells expand in thyroid tissue with ICI treatment. Intrathyroidal IL-17A-producing innate-like γδT17 cells were increased in tumor-free mice, whereas adaptive Th17 cells were also prominent in tumor-bearing mice, following ICI treatment. Furthermore, Ab-based inhibition of IL-17A, a clinically available therapy, significantly reduced thyroid IrAE development in ICI-treated mice with and without tumor challenge. Finally, combination of IL-17A neutralization with ICI treatment in multiple tumor models did not reduce ICI antitumor efficacy. These studies suggest that targeting Th17 and γδT17 cell function via the IL-17A axis may reduce IrAEs without impairing ICI antitumor efficacy and may be a generalizable strategy to address type 3 immune-mediated IrAEs.
Collapse
Affiliation(s)
- Melissa G Lechner
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA;
| | - Mandy I Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Anushi Y Patel
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Aline T Hoang
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA
| | | | - Michael Astourian
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Marissa S Pioso
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Eduardo D Rodriguez
- Department of Pathology, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Ethan C McCarthy
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Willy Hugo
- Division of Dermatology, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Trevor E Angell
- Division of Endocrinology and Diabetes, USC Keck School of Medicine, Los Angeles, CA
| | - Alexandra Drakaki
- Division of Hematology and Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA; and
| | - Antoni Ribas
- Division of Hematology and Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA; and
| | - Maureen A Su
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA
- Division of Pediatric Endocrinology, UCLA David Geffen School of Medicine, Los Angeles, CA
| |
Collapse
|