1
|
van Dijk B, Janssen JC, van Daele PLA, de Jonge MJA, Joosse A, Verheul HMW, Epker JL, van der Veldt AAM. From ICI to ICU: A systematic review of patients with solid tumors who are treated with immune checkpoint inhibitors (ICI) and admitted to the intensive care unit (ICU). Cancer Treat Rev 2025; 136:102936. [PMID: 40222269 DOI: 10.1016/j.ctrv.2025.102936] [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: 02/18/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) have improved the survival of patients with different solid tumors and even resulted in cure of metastatic disease. Since the introduction of ICIs, an increasing number of patients is admitted to the ICU for severe and potentially life-threatening immune related adverse events (irAEs). The outcome of patients who are admitted to the ICU because of severe irAEs is still unknown. The aim of this systematic review is to collect evidence on the outcomes of patients with solid tumors who are admitted to the ICU because of irAEs. METHODS Medline, Embase, Cochrane central register of controlled trials and Google Scholar were searched systematically from 1975 to 24 September 2024. Articles were only included when describing patients with solid tumors who were admitted to the ICU because of irAEs after treatment with ICIs. Two independent reviewers extracted the data and assessed the risk of bias. RESULTS A total of 183 articles were included: two prospective ICU population-based studies, four retrospective ICU population-based studies, 25 retrospective studies describing irAEs with incidental ICU admissions, one review of case reports, and 153 articles with a total of 177 case reports. The six ICU population-based studies contained a total of 169 patients who were admitted to the ICU due to irAEs. In these six studies, the most frequently reported irAEs were pneumonitis and neurological irAEs. Of these 169 patients, 26% of the patients died on the ICU and an additional 8% of patients in the three to six months thereafter due to irAEs or disease progression. In all 183 included articles, various irAEs were described and the reported mortality rate varied from 0 to 53%. CONCLUSION The potential favorable outcomes of both the solid tumors and irAEs will probably result in more need for ICU admissions. Prospective clinical trials are needed to optimize the treatment strategy of severe irAEs at the ICU. Based on the favourable outcomes after life-threatening irAEs, ICU admission should definitely be considered for patients with solid tumors who have life-threatening irAEs.
Collapse
Affiliation(s)
- Brigit van Dijk
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Joséphine C Janssen
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands; Department of Oncological Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Paul L A van Daele
- Department of Clinical Immunology, Erasmus MC, Rotterdam, the Netherlands
| | - Maja J A de Jonge
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Jelle L Epker
- Department of Intensive Care, Erasmus MC, Rotterdam, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
2
|
Ikeda H. Cancer immunotherapy in progress-an overview of the past 130 years. Int Immunol 2025; 37:253-260. [PMID: 39792088 PMCID: PMC11975553 DOI: 10.1093/intimm/dxaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/09/2025] [Indexed: 01/12/2025] Open
Abstract
Since the first approval of an immune checkpoint inhibitor, we have witnessed the clinical success of cancer immunotherapy. Adoptive T-cell therapy with chimeric antigen receptor T (CAR-T) cells has shown remarkable efficacy in hematological malignancies. Concurrently with these successes, the cancer immunoediting concept that refined the cancer immunosurveillance concept underpinned the scientific mechanism and reason for past failures, as well as recent breakthroughs in cancer immunotherapy. Now, we face the next step of issues to be solved in this field, such as tumor heterogeneity, the tumor microenvironment, the metabolism of tumors and the immune system, and personalized approaches for patients, aiming to expand the population benefitted by the therapies.
Collapse
Affiliation(s)
- Hiroaki Ikeda
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
3
|
Williamson J, Fadlullah MZH, Kovacsovics-Bankowski M, Gibson B, Swami U, Erickson-Wayman A, Jamison D, Sageser D, Jeter J, Bowles TL, Cannon DM, Haaland B, Schroeder JD, Nix DA, Atkinson A, Hyngstrom J, McPherson J, Tan AC, Hu-Lieskovan S. Dramatic Responses to High-Dose Ipilimumab Plus Temozolomide After Progression on Standard- or Low-Dose Ipilimumab in Advanced Melanoma. Curr Oncol 2025; 32:144. [PMID: 40136348 PMCID: PMC11941404 DOI: 10.3390/curroncol32030144] [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: 10/29/2024] [Revised: 02/10/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Patients with advanced melanoma who progress on standard-dose ipilimumab (Ipi) + nivolumab continue to have poor prognosis. Studies support a dose-response activity of Ipi, and one promising combination is Ipi 10 mg/kg (Ipi10) + temozolomide (TMZ). We performed a retrospective cohort analysis of patients with advanced melanoma treated with Ipi10 + TMZ in the immunotherapy refractory/resistant setting (n = 6, all progressed after prior Ipi + nivolumab), using similar patients treated with Ipi3 + TMZ (n = 6) as comparison. Molecular profiling by whole-exome sequencing (WES) and RNA-sequencing (RNA-seq) of tumors harvested through one responder's treatment was performed. With a median follow up of 119 days, patients treated with Ipi10 + TMZ had a statistically significant longer median progression-free survival of 144.5 days (range 27-219) vs. 44 (26-75) in Ipi 3 mg/kg (Ipi3) + TMZ, p = 0.04, and a trend of longer median overall survival of 154.5 days (27-537) vs. 89.5 (26-548). Two patients in the Ipi10 + TMZ cohort had a partial response, and both responders had BRAF V600E mutant melanoma. RNA-seq showed enrichment of inflammatory signatures, including interferon responses in metastases after Ipi10 + TMZ compared to the primary tumor, and downregulated negative immune regulators. Ipi10 + TMZ demonstrated efficacy, including dramatic responses in patients refractory to prior Ipi + anti-PD1. Molecular data suggest a potential threshold of Ipi dose for activation of sufficient anti-tumor immune response, and higher doses are required for some patients.
Collapse
Affiliation(s)
- Julie Williamson
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT 84112, USA; (J.W.)
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | | | | | - Berit Gibson
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Umang Swami
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT 84112, USA; (J.W.)
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Alyssa Erickson-Wayman
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Debra Jamison
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Dan Sageser
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Joanne Jeter
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT 84112, USA; (J.W.)
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | | | - Donald M. Cannon
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Ben Haaland
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Joyce D. Schroeder
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - David A. Nix
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Aaron Atkinson
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - John Hyngstrom
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Jordan McPherson
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Aik-Choon Tan
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| | - Siwen Hu-Lieskovan
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT 84112, USA; (J.W.)
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA (B.G.); (D.S.); (D.M.C.); (A.A.); (J.H.)
| |
Collapse
|
4
|
Connor C, Carr QL, Sweazy A, McMasters K, Hao H. Clinical Approaches for the Management of Skin Cancer: A Review of Current Progress in Diagnosis, Treatment, and Prognosis for Patients with Melanoma. Cancers (Basel) 2025; 17:707. [PMID: 40002300 PMCID: PMC11853469 DOI: 10.3390/cancers17040707] [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: 01/20/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Melanoma represents a significant public health challenge due to its increasing incidence and potential for metastasis. This review will explore the current clinical approaches to the management of melanoma, focusing on advancements in diagnosis, treatment, and prognosis. Methods for early detection and accurate staging have been enhanced by new diagnostic strategies. Treatment modalities have expanded beyond traditional surgical excision to include targeted therapy and immunotherapy. Prognostic assessment has benefited from the development of novel biomarkers and genetic profiling. This review will highlight the progress made in the multidisciplinary management of melanoma, underscoring the importance of continuous research to improve patient outcomes.
Collapse
Affiliation(s)
- Colton Connor
- School of Medicine, University of Louisville, Louisville, KY 40202, USA; (C.C.); (Q.L.C.)
| | - Quinton L. Carr
- School of Medicine, University of Louisville, Louisville, KY 40202, USA; (C.C.); (Q.L.C.)
| | - Alisa Sweazy
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| | - Kelly McMasters
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| | - Hongying Hao
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| |
Collapse
|
5
|
Li J, Zhou X, Wu L, Ma J, Tan Y, Wu S, Zhu J, Wang Q, Shi Q. Optimal early endpoint for second-line or subsequent immune checkpoint inhibitors in previously treated advanced solid cancers: a systematic review. BMC Cancer 2025; 25:293. [PMID: 39966752 PMCID: PMC11837729 DOI: 10.1186/s12885-025-13712-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The administration of second-line or subsequent immune checkpoint inhibitors (ICIs) in previously treated patients with advanced solid cancers has been clinically investigated. However, previous clinical trials lacked an appropriate primary endpoint for efficacy assessment. This systematic review aimed to explore the most optimal early efficacy endpoint for such trials. METHODS Phase 2 or 3 clinical trials involving patients with advanced solid cancers with disease progression following standard first-line therapy receiving second-line or subsequent ICI administration, with adequate survival outcome data, were included from PubMed, Embase, Web of Science, and Cochrane Library databases before February 2023. Quality assessment was conducted using the Cochrane tool and Newcastle-Ottawa Quality Assessment Scale for Cohort Studies for randomized controlled trials (RCTs) and non-randomized trials, respectively. Objective response rate (ORR) and progression-free survival (PFS) at 3, 6, and 9 months were investigated as potential early efficacy endpoint candidates for 12-month overall survival (OS), with a strong correlation defined as Pearson's correlation coefficient r ≥ 0.8. RESULTS A total of 64 RCTs comprising 22,725 patients and 106 non-randomized prospective trials involving 10,608 participants were eligible for modeling and external validation, respectively. RCTs examined 15 different cancer types, predominantly non-small-cell lung cancer (NSCLC) (17, 28%), melanoma (9, 14%), and esophageal squamous cell carcinoma (5, 8%). The median sample size of RCTs was 124 patients, and the median follow-up time was 3.2-57.7 months. The ORR (r = 0.38; 95% confidence interval [CI], 0.18-0.54) and PFS (r = 0.42; 95% CI, 0.14-0.64) exhibited weak trial-level correlations with OS. Within ICI treatment arms, the r values of ORR and 3-, 6-, and 9-month PFS with 12-month OS were 0.61 (95% CI, 0.37-0.79), 0.78 (95% CI, 0.62-0.88), 0.84 (95% CI, 0.77-0.90), and 0.86 (95% CI, 0.79-0.90), respectively. External validation of 6-month PFS indicated an acceptable discrepancy between actual and predicted 12-month OS. CONCLUSIONS In non-randomized phase 2 trials on second-line or subsequent ICI therapy in patients with advanced solid cancers, 6-month PFS could serve as an early efficacy endpoint. However, early efficacy endpoints are not recommended in RCTs to replace OS.
Collapse
Affiliation(s)
- Jingqiu Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoding Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Wu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiabao Ma
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Tan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Songke Wu
- Department of Oncology, People'S Hospital of Cangxi County, Guangyuan, China.
| | - Jie Zhu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qiuling Shi
- Center for Cancer Prevention Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Hu Y, Yang R, Ni S, Song Z. Bibliometric analysis of targeted immunotherapy for osteosarcoma-current knowledge, hotspots and future perspectives. Front Immunol 2025; 15:1485053. [PMID: 39995821 PMCID: PMC11847827 DOI: 10.3389/fimmu.2024.1485053] [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: 08/23/2024] [Accepted: 12/24/2024] [Indexed: 02/26/2025] Open
Abstract
Objective The objective of this study is to conduct a bibliometric analysis on examining the current condition, areas of interest, and rising trends of immunotherapy for osteosarcoma (ITFOS), as well as its importance in associated research domains. Methods An extensive collection of academic papers on the use of ITFOS was obtained from the Web of Science between January 1, 2000 and October 20, 2023. Then, using a variety of tools like HisCite, VOSviewer, CiteSpace, and the bibliometrix package, a bibliometric study was carried out. This study included the collection of information on country, institution, author, journal, and keywords. Results A comprehensive analysis was undertaken on a total of 616 publications obtained from 247 journals, encompassing the contributions of 3725 authors affiliated with 831 institutes spanning across 43 countries/regions. Notably, China exhibited the highest quantity of published 277 (44.99%) articles on ITFOS. The most productive institution was Zhejiang University, with 26 (4.22%) publications. The author with the highest publication output was Tsukahara, Tomohide from Japan with 15 (2.44%) publications. The article with the most citation was "DOI: 10.1200/JCO.2014.58.0225". Frontiers in Immunology demonstrated the highest level of productivity, having published a total of 31 (5.03%) articles. The most frequently used were "osteosarcoma," "immunotherapy," and "cancer,". Meanwhile, "sequencing", "prognostic signature" and "immune microenvironment" have been identified as the research frontiers for the forthcoming years. Conclusion This paper provides a thorough evaluation of current research trends and advancements in ITFOS. It includes relevant research findings and emphasizes collaborative efforts among authors, institutions, and countries.
Collapse
Affiliation(s)
- Yunxiang Hu
- Department of Orthopaedic Trauma, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, China
| | - Rui Yang
- School of Graduates, Dalian Medical University, Dalian, Liaoning, China
| | - Shuai Ni
- Department of Orthopaedic Trauma, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, China
| | - Zefeng Song
- School of Graduates, Dalian University of Technology, Dalian, Liaoning, China
| |
Collapse
|
7
|
Kim TK, Lee HS, Kim ES. Incidence and risk factors of immune checkpoint inhibitor-induced colitis in Korean patients with cancer. Korean J Intern Med 2025; 40:49-56. [PMID: 39778525 PMCID: PMC11725480 DOI: 10.3904/kjim.2024.135] [Citation(s) in RCA: 1] [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: 04/21/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/AIMS Immune checkpoint inhibitors (ICIs) are effective in treating cancer. However, various immune-related adverse events (irAEs) have become prevalent, with ICI-induced colitis being the most common gastrointestinal irAE. Thus, we aimed to investigate the incidence and risk factors of ICI-induced colitis in Korean patients with cancer. METHODS This retrospective study included patients treated with ICIs between October 2015 and June 2022 in two tertiary referral centers in Daegu, Korea. The incidence of ICI-induced colitis was determined using electronic medical records. Risk factors for ICI-induced colitis were identified using univariate and multivariate logistic regression analyses. RESULTS We included 1,478 patients with ICI-treated cancer. The incidence of ICI-induced colitis was 3.5% (n = 52/1,478). Multivariate logistic regression analysis showed that the combination of nivolumab and ipilimumab was a risk factor for ICI-induced colitis (p = 0.006; odds ratio, 9.768; 95% confidence interval, 1.93-49.30). CONCLUSION ICI-induced colitis had an incidence rate of 3.5% and was associated with the combination of nivolumab and ipilimumab. Most patients with ICI-induced colitis developed mild symptoms that improved with supportive care alone, making ICI therapy resumption possible.
Collapse
Affiliation(s)
- Tae Kyun Kim
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu,
Korea
| | - Hyun Seok Lee
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu,
Korea
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu,
Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Eun Soo Kim
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu,
Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu,
Korea
| |
Collapse
|
8
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
9
|
Zhang L, Maalouf A, Makri SC, Banerjee J, Suru A, Tam AJ, Calizo A, Pollard K, Wang J, Danilova L, Ioannou M, Levin AS, Morris CD, Rhee DS, Belzberg AJ, Blakeley JO, Ladle BH, Pardoll DM, Lucas CHG, Rodriguez FJ, Gross JM, Anders RA, Pratilas CA, Llosa NJ. Multidimensional Immunotyping of Human NF1-Associated Peripheral Nerve Sheath Tumors Uncovers Tumor-Associated Macrophages as Key Drivers of Immune Evasion in the Tumor Microenvironment. Clin Cancer Res 2024; 30:5459-5472. [PMID: 39321200 PMCID: PMC11866061 DOI: 10.1158/1078-0432.ccr-24-1454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/15/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
PURPOSE Malignant peripheral nerve sheath tumors (MPNST) are aggressive soft-tissue sarcomas and the leading cause of mortality in individuals with neurofibromatosis type 1 (NF1). Despite many clinical trials, outcomes for patients with MPNST have remained stagnant, and most succumb to their disease; thus, novel therapeutic approaches are needed. A better understanding of the MPNST immune ecosystem will aid in the development of strategies to activate the immune system against the tumor. In this study, we profile the tumor immune microenvironment (TIME) in NF1-associated peripheral nerve sheath tumors (PNST) to discover insights on the role played by tumor-infiltrating immune cells in malignant transformation. EXPERIMENTAL DESIGN Using fresh and formalin-fixed paraffin-embedded tissue from patients diagnosed with NF1-PNST, we dissected the TIME through IHC, multiparameter flow cytometry, and comparative transcriptomic studies. RESULTS Immunophenotyping confirmed increased immune cell infiltration during malignant progression, with a predominance of infiltrating myeloid cells, particularly CD163+ tumor-associated macrophages (TAM). The T cells within MPNST exhibited signs of tumor activation, characterized by high programmed cell death 1 expression. Additionally, MPNST specimens demonstrated elevated levels of immunosuppressive TAM, with heightened PD-L1 expression. The proportion of CD163+ myeloid cells within the TIME correlated with poorer progression-free survival. Notably, loss of H3K27 trimethylation correlated with low immune cell infiltration in MPNST. CONCLUSIONS Malignant transformation of NF1-PNST is characterized by an immunosuppressive microenvironment comprising TAM with high expression of PD-L1, which is associated with inferior outcomes. These findings suggest the clinical potential of immune-modulating therapeutics that can unleash an antitumor immune response.
Collapse
MESH Headings
- Humans
- Tumor Microenvironment/immunology
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
- Tumor-Associated Macrophages/pathology
- Neurofibromatosis 1/immunology
- Neurofibromatosis 1/pathology
- Neurofibromatosis 1/genetics
- Neurofibromatosis 1/complications
- Immunophenotyping
- Nerve Sheath Neoplasms/pathology
- Nerve Sheath Neoplasms/immunology
- Nerve Sheath Neoplasms/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Tumor Escape
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Neurofibrosarcoma/pathology
- Neurofibrosarcoma/genetics
- Neurofibrosarcoma/immunology
- Female
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Male
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- CD163 Antigen
Collapse
Affiliation(s)
- Lindy Zhang
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Alexandre Maalouf
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Stavriani C. Makri
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Aditya Suru
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Ada J. Tam
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Ana Calizo
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kai Pollard
- Laboratory Corporation of America (Labcorp), Burlington, NC
| | - Jiawan Wang
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ludmila Danilova
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Maria Ioannou
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Adam S. Levin
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Carol D. Morris
- Orthopedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel S. Rhee
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Allan J. Belzberg
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jaishri O. Blakeley
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Brian H. Ladle
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Drew M. Pardoll
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | | | - Fausto J. Rodriguez
- Department of Pathology, University of California Los Angeles, Los Angeles, CA
| | - John M. Gross
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Robert A. Anders
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Christine A. Pratilas
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nicolas J. Llosa
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
10
|
Idowu O, Lewis A, Doyle CA. Perioperative Implications of Biologics and Immunotherapy. Adv Anesth 2024; 42:97-113. [PMID: 39443053 DOI: 10.1016/j.aan.2024.07.008] [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: 10/25/2024]
Abstract
Immune check inhibitors (ICIs) are a class of biologic therapy used for cancer treatment that enhances T-cell recognition of cancer cells. Toxicities from ICIs are described as immune-related adverse events (irAEs) with Grade 1 to 2 irAEs representing mild-to-moderate toxicity and Grade 3 to 4 irAEs representing severe to life-threatening toxicity. The long half-life of ICIs contributes to the extended and unpredictable nature of irAEs. ICI therapy is typically stopped for Grade 3 to 4 irAEs except for endocrinopathies if clinically optimized. Toxicities can involve any organ system; therefore, a thorough preoperative assessment is imperative to ensure appropriate clinical management.
Collapse
Affiliation(s)
- Olakunle Idowu
- Department of Anesthesiology & Perioperative Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA.
| | - Alexandra Lewis
- Department of Anesthesiology & Critical Care Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, M-316, New York, NY 10065, USA
| | - Christine Anne Doyle
- Department of Anesthesiology, O'Connor Hospital, 2077 Walnut Grove Avenue, San Jose, CA 95128, USA
| |
Collapse
|
11
|
Wang R, Zhu XY, Wang Y. Knowledge graph and frontier trends in melanoma-associated ncRNAs: a bibliometric analysis from 2006 to 2023. Front Oncol 2024; 14:1439324. [PMID: 39659781 PMCID: PMC11628868 DOI: 10.3389/fonc.2024.1439324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/24/2024] [Indexed: 12/12/2024] Open
Abstract
Objectives Malignant melanoma (MM) is a highly malignant skin tumor. Although research on non-coding RNAs (ncRNAs) of MM has advanced swiftly in recent years, no specific bibliometric analyses have been conducted on this topic. The present study aims to summarize the knowledge graphs and frontier trends and to provide new perspectives and direction of collaboration for researchers. Method Research data on melanoma and ncRNA published from January 1, 2006 to October 9, 2023 were retrieved and extracted from Web of Science. R Studio (Version 4.3.1), Scimago Graphica (Version 1.0.36), VOSviewer version (1.6.19), and Citespace (6.2.4R) were used to analyze the publications, countries, journals, institutions, authors, keywords, references, and other relevant data and to build collaboration network graphs and co-occurrence network graphs accordingly. Results A total of 1,222 articles were retrieved, involving 4,894 authors, 385 journals, 43,220 references, 2413 keywords, and 1,651 institutions in 47 countries. The average annual growth rate in the number of articles was 25.02% from 2006 to 2023; among all the journals, Plos One had the highest number of publications and citations, which are 42 publications and 2,228 citations, respectively. Chinese researchers were the most prolific publishers in this field, having published a total of 657 articles, among which 42 were published by Shanghai Jiao Tong University, which was the most productive institution. In recent years, the most explored keywords included long non-coding RNAs, immunotherapy, and exosm. According to the timeline chart of reference co-citation, "functional role" has been the most explored hotspot since 2015, and human cancer is a newly emerged hotspot after 2021. Conclusion Through a bibliometric analysis, this study included all publications on ncRNAs and melanoma that were published in English from 2006 to 2023 in Web of Science to analyze the trends in the number of publications, international research focuses, and the direction of collaboration. The results of this study may provide information on knowledge graph, frontier trends and identify research topics in melanoma. More current research proved that ncRNA plays a crucial role in the biological behavior of melanoma including proliferation, invasion, metastasis, drug resistance, etc. With the development of research on ncRNA and melanoma, ncRNA may great potential in development of early diagnosis, targeted therapy and efficacy evaluation in the future. The results of this study also provide new perspectives and research partners for researchers in this field.
Collapse
Affiliation(s)
- Ru Wang
- Department of Pediatrics, Xinzhou District People’s Hospital, Wuhan, Hubei, China
| | - Xiao-yan Zhu
- Sanquan College of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yi Wang
- The Fifth People’s Hospital of Hainan Province, Affiliated Dermatology Hospital of Hainan Medical University, Haikou, Hainan, China
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Ngo HX, Oh E, Li C, Yu J. Oncology Dose Selection in Subsequent Indications: What Can We Learn From FDA-approved Oncology Drugs? Clin Ther 2024; 46:927-937. [PMID: 39304367 DOI: 10.1016/j.clinthera.2024.08.020] [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: 06/28/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE The modern oncology drug development landscape has shifted away from traditional cytotoxic chemotherapies. Following their initial approvals, many oncology drugs have been approved in subsequent indications either as monotherapy or in combination to benefit a broader patient population. To date, dose selection strategies for subsequent indications have not been systematically reviewed. This review examines how approved dosing regimens were selected in subsequent indications for FDA-approved oncology drugs. METHODS The Drugs@FDA database was used to identify FDA-approved new molecular entities (NMEs) between 2010 and 2023. NMEs with more than 1 approved indication were included in the analysis. In total, the dosing regimens for 67 novel oncology drugs that obtained FDA approvals for multiple indications were evaluated. FINDINGS Overall, in subsequent indications, 72% of NMEs used the same or clinically equivalent alternative dosing regimens to those approved in the initial indications. Amongst the 28% of NMEs that used different dosing regimens, safety/tolerability was the leading cause of a dosing regimen changes in both monotherapy and combination therapy settings. Other factors leading to changes in dosing regimens include differences in tumor biology, disease burden, pharmacokinetics, and overall benefit-risk profiles obtained from dose-finding studies. IMPLICATIONS Our analysis highlighted the importance of selecting a safe, tolerable, and yet efficacious dosing regimen for the initial indication as a suboptimal initially approved regimen could lead to dosing regimen changes in later indications. Preclinical and clinical data could be leveraged to understand the pharmacology, pharmacokinetic, and pharmacodynamic differences between indications and thus support dose selection in subsequent indications.
Collapse
Affiliation(s)
- Huy X Ngo
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Elise Oh
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Chunze Li
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Jiajie Yu
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA.
| |
Collapse
|
13
|
Tsimberidou AM, Alayli FA, Okrah K, Drakaki A, Khalil DN, Kummar S, Khan SA, Hodi FS, Oh DY, Cabanski CR, Gautam S, Meier SL, Amouzgar M, Pfeiffer SM, Kageyama R, Yang E, Spasic M, Tetzlaff MT, Foo WC, Hollmann TJ, Li Y, Adamow M, Wong P, Moore JS, Velichko S, Chen RO, Kumar D, Bucktrout S, Ibrahim R, Dugan U, Salvador L, Hubbard-Lucey VM, O’Donnell-Tormey J, Santulli-Marotto S, Butterfield LH, Da Silva DM, Fairchild J, LaVallee TM, Padrón LJ, Sharma P. Immunologic signatures of response and resistance to nivolumab with ipilimumab in advanced metastatic cancer. J Exp Med 2024; 221:e20240152. [PMID: 39190534 PMCID: PMC11349049 DOI: 10.1084/jem.20240152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/13/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
Identifying pan-tumor biomarkers that predict responses to immune checkpoint inhibitors (ICI) is critically needed. In the AMADEUS clinical trial (NCT03651271), patients with various advanced solid tumors were assessed for changes in intratumoral CD8 percentages and their response to ICI. Patients were grouped based on tumoral CD8 levels: those with CD8 <15% (CD8-low) received nivolumab (anti-PD-1) plus ipilimumab (anti-CTLA4) and those with CD8 ≥15% (CD8-high) received nivolumab monotherapy. 79 patients (72 CD8-low and 7 CD8-high) were treated. The disease control rate was 25.0% (18/72; 95% CI: 15.8-35.2) in CD8-low and 14.3% (1/7; 95% CI: 1.1-43.8) in CD8-high. Tumors from 35.9% (14/39; 95% CI: 21.8-51.4) of patients converted from CD8 <15% pretreatment to ≥15% after treatment. Multiomic analyses showed that CD8-low responders had an inflammatory tumor microenvironment pretreatment, enhanced by an influx of CD8 T cells, CD4 T cells, B cells, and macrophages upon treatment. These findings reveal crucial pan-cancer immunological features for ICI response in patients with metastatic disease.
Collapse
Affiliation(s)
- Apostolia M. Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farah A. Alayli
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Kwame Okrah
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | | | | | | | - F. Stephen Hodi
- Dana-Farber Cancer Institute, Boston, MA, USA
- Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Y. Oh
- University of California, San Francisco, San Francisco, CA, USA
| | | | - Shikha Gautam
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Stefanie L. Meier
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Stanford University, Stanford, CA, USA
| | - Meelad Amouzgar
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Robin Kageyama
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - EnJun Yang
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Marko Spasic
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Michael T. Tetzlaff
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wai Chin Foo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Travis J. Hollmann
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Bristol Myers Squibb, New York, NY, USA
| | - Yanyun Li
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew Adamow
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Phillip Wong
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Dinesh Kumar
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Ute Dugan
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | | | | | | | | | | | - Justin Fairchild
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Lacey J. Padrón
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Padmanee Sharma
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Immunotherapy Platform, James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
Moscardini-Martelli J, Rodríguez-Camacho A, Torres-Ríos JA, Meraz-Soto JM, Flores-Vázquez JG, Hernández-Sánchez LC, Lozano-Ruiz FJ, Maldonado-Magos F, Cid-Sánchez D, Flores-Balcázar CH, Celis-López MÁ, Gutiérrez-Aceves GA, Flores-Vázquez F, Moreno-Jiménez S. A Comprehensive Revision of Radiation Immunotherapy and the Abscopal Effect in Central Nervous System Metastases: Reassessing the Frontier. Curr Issues Mol Biol 2024; 46:11075-11085. [PMID: 39451538 PMCID: PMC11506806 DOI: 10.3390/cimb46100658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024] Open
Abstract
Seventy years ago, Robin Mole introduced the concept of the abscopal effect to describe a rare phenomenon. This occurs when local radiation triggers an immune-mediated reduction in tumors outside the treated area but within the same organism. Observing this effect has been linked to improved overall and progression-free survival in patients who experience it. While the abscopal effect was once considered rare, it is now being observed more frequently due to the combination of radiation with immunotherapy. As a result, more researchers are exploring this study area, which shows promise for excellent results. This review focuses explicitly on the immunological implications of activating the abscopal effect through ionizing radiation in the central nervous system and explores the potentially involved immunological pathways.
Collapse
Affiliation(s)
- Júlia Moscardini-Martelli
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Alejandro Rodríguez-Camacho
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Jorge Alejandro Torres-Ríos
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Juan Marcos Meraz-Soto
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - José Guillermo Flores-Vázquez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Laura Crystell Hernández-Sánchez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Francisco Javier Lozano-Ruiz
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Federico Maldonado-Magos
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Dharely Cid-Sánchez
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | | | - Miguel Ángel Celis-López
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Guillermo Axayacatl Gutiérrez-Aceves
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | | | - Sergio Moreno-Jiménez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
- American British Cowdray Medical Center, Neurological Center, Mexico City 01120, Mexico
| |
Collapse
|
15
|
Luo Y, Lu J, Lei Z, Zhu H, Rao D, Wang T, Fu C, Zhang Z, Xia L, Huang W. Lysine methylation modifications in tumor immunomodulation and immunotherapy: regulatory mechanisms and perspectives. Biomark Res 2024; 12:74. [PMID: 39080807 PMCID: PMC11289998 DOI: 10.1186/s40364-024-00621-w] [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: 06/11/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024] Open
Abstract
Lysine methylation is a crucial post-translational modification (PTM) that significantly impacts gene expression regulation. This modification not only influences cancer development directly but also has significant implications for the immune system. Lysine methylation modulates immune cell functions and shapes the anti-tumor immune response, highlighting its dual role in both tumor progression and immune regulation. In this review, we provide a comprehensive overview of the intrinsic role of lysine methylation in the activation and function of immune cells, detailing how these modifications affect cellular processes and signaling pathways. We delve into the mechanisms by which lysine methylation contributes to tumor immune evasion, allowing cancer cells to escape immune surveillance and thrive. Furthermore, we discuss the therapeutic potential of targeting lysine methylation in cancer immunotherapy. Emerging strategies, such as immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T-cell (CAR-T) therapy, are being explored for their efficacy in modulating lysine methylation to enhance anti-tumor immune responses. By targeting these modifications, we can potentially improve the effectiveness of existing treatments and develop novel therapeutic approaches to combat cancer more effectively.
Collapse
Affiliation(s)
- Yiming Luo
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Junli Lu
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhen Lei
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - He Zhu
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dean Rao
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Tiantian Wang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chenan Fu
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhiwei Zhang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Wenjie Huang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China.
| |
Collapse
|
16
|
Saowapa S, Polpichai N, Tanariyakul M, Wannaphut C, Wattanachayakul P, Danpanichkul P, Suenghataiphorn T, Kulthamrongsri N, Siladech P, Tijani L. Immunotherapy-induced hepatitis in metastatic colorectal cancer: a systematic review and meta-analysis. Proc AMIA Symp 2024; 37:841-850. [PMID: 39165807 PMCID: PMC11332647 DOI: 10.1080/08998280.2024.2374161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/12/2024] [Accepted: 06/16/2024] [Indexed: 08/22/2024] Open
Abstract
Recent advances in immunotherapy using immune checkpoint inhibitors (ICIs) for various cancers have also highlighted a rise in immune-related adverse events, including hepatitis, potentially leading to the discontinuation of treatment. This study aimed to evaluate the prevalence of hepatitis in metastatic colorectal cancer (mCRC) patients undergoing different ICI therapies. An extensive search of PubMed, PubMed Central, and Google Scholar up to November 2023 identified relevant studies. After excluding non-English articles, case reports, reviews, ongoing trials, and studies combining other therapies, five studies qualified for inclusion. Data extraction and statistical analyses were performed using Excel and Comprehensive Meta-Analysis software, respectively. Results from a subgroup analysis indicated that the incidence of hepatitis was comparable among patients treated with PD-1 monotherapy, PDL-1 monotherapy, and combination PD-1 and CTLA-4 therapy, with rates of 2.6%, 2.2%, and 1.7% for any grade and 2.1%, 2.2%, and 1.7% for grade ≥3 hepatitis, respectively. Naive-treated mCRC patients exhibited higher hepatitis rates than those previously treated (3.2% vs 1.6% and 2.6% vs 1.6% for any grade and grade ≥3, respectively). This study underscores the similar risk of hepatitis across different ICI therapies, with an increased incidence in naive-treated mCRC patients.
Collapse
Affiliation(s)
- Sakditad Saowapa
- Department of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Natchaya Polpichai
- Department of Internal Medicine, Weiss Memorial Hospital, Chicago, Illinois, USA
| | - Manasawee Tanariyakul
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Chalothorn Wannaphut
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | | | - Pojsakorn Danpanichkul
- Department of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | | | - Narathorn Kulthamrongsri
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Pharit Siladech
- Department of Internal Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Lukman Tijani
- Hematology and Oncology Department, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
17
|
Younis A, Gribben J. Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions. IMMUNO 2024; 4:186-210. [DOI: 10.3390/immuno4030013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) are a promising form of immunotherapy that have significantly changed the therapeutic landscape for many advanced cancers. They have shown unique clinical benefit against a broad range of tumour types and a strong overall impact on survival in studied patient populations. However, there are still many limitations holding back this immunotherapy from reaching its full potential as a possible curative option for advanced cancer patients. A great deal of research is being undertaken in the hope of driving advancements in this area, building a better understanding of the mechanisms behind immune checkpoint inhibition and ultimately developing more effective, safer, and wider-reaching agents. Taking into account the current literature on this topic, this review aims to explore in depth the basis of the use of ICIs in the treatment of advanced cancers, evaluate its efficacy and safety, consider its current limitations, and finally reflect on what the future holds for this very promising form of cancer immunotherapy.
Collapse
Affiliation(s)
- Abdullah Younis
- Barts and the London School of Medicine and Dentistry, London E1 2AD, UK
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6AU, UK
| |
Collapse
|
18
|
Wojciechowicz K, Kuncewicz K, Rutkowski J, Jassem J, Rodziewicz-Motowidło S, Wardowska A, Spodzieja M. Targeting BTLA with the peptide inhibitor HVEM(14-39) - A new way to restore the activity of T cells in melanoma. Biomed Pharmacother 2024; 175:116675. [PMID: 38733770 DOI: 10.1016/j.biopha.2024.116675] [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: 03/01/2024] [Revised: 04/21/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
The complex of B- and T-lymphocyte attenuator (BTLA) and herpes virus entry mediator (HVEM) plays a critical role in immune regulation and has emerged as a promising therapeutic target for cancer treatment. In this study, we investigated the potential of the peptide inhibitor HVEM(14-39) to restore peripheral T cell activity in patients with advanced melanoma. In these patients, CD8+ T cells downregulated BTLA expression and increased HVEM expression upon activation. The addition of HVEM(14-39) reduced the percentage of BTLA+ CD8+ T cells and increased the subpopulation of HVEM+ CD8+ T cells. Additionally, HVEM(14-39) enhanced T cell activation, proliferation, and the shift toward effector memory T cell subpopulations. Finally, this peptide affected the proliferation rate and late apoptosis of melanoma cell line in co-culture with T cells. These findings suggest that HVEM(14-39) can overcome T cell exhaustion and improve antitumor responses. Peptide-based immunotherapy targeting the BTLA-HVEM complex offers a promising alternative to monoclonal antibody-based therapies, with the potential for fewer side effects and higher treatment efficacy.
Collapse
Affiliation(s)
- Karolina Wojciechowicz
- Department of Physiopathology, Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Katarzyna Kuncewicz
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Jacek Rutkowski
- Department of Oncology and Radiotherapy, Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Jacek Jassem
- Department of Oncology and Radiotherapy, Faculty of Medicine, Medical University of Gdańsk, Poland
| | | | - Anna Wardowska
- Department of Physiopathology, Faculty of Medicine, Medical University of Gdańsk, Poland.
| | - Marta Spodzieja
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland.
| |
Collapse
|
19
|
Miwa S, Hayashi K, Taniguchi Y, Asano Y, Demura S. What are the Optimal Systemic Treatment Options for Rhabdomyosarcoma? Curr Treat Options Oncol 2024; 25:784-797. [PMID: 38750399 DOI: 10.1007/s11864-024-01206-3] [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: 04/14/2024] [Indexed: 07/04/2024]
Abstract
OPINION STATEMENT Rhabdomyosarcoma, a soft tissue sarcoma commonly observed in childhood, requires multidisciplinary treatment, including surgical tumor resection, chemotherapy, and radiation therapy. Although long-term survival can be expected in patients with localized rhabdomyosarcoma, the clinical outcomes in patients with metastatic or unresectable rhabdomyosarcoma remain unsatisfactory. To improve the outcomes of rhabdomyosarcoma, it is important to explore effective systemic treatments for metastatic rhabdomyosarcoma. Currently, multiagent chemotherapy comprising vincristine, actinomycin D, and ifosfamide/cyclophosphamide remains standard systemic treatment for rhabdomyosarcoma. On the other hand, new treatment, such as immune checkpoint inhibitors and molecular targeted drugs, have demonstrated superior clinical outcomes compared to those of standard treatments in various type of malignancies. Therefore, it is necessary to assess the efficacies of these treatments in patients with rhabdomyosarcoma. Recent clinical studies have shown efficacies and safeties of temozolomide combined with vincristine/irinotecan, olaratumab combined with doxorubicin or vincristine/irinotecan, and long-term maintenance therapy. Furthermore, basic researches demonstrated new therapeutic targets. Future studies using these approaches are required to assess their clinical significances.
Collapse
Affiliation(s)
- Shinji Miwa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan.
| | - Katsuhiro Hayashi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Yuta Taniguchi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Yohei Asano
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Satoru Demura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| |
Collapse
|
20
|
Haugh A, Daud AI. Therapeutic Strategies in BRAF V600 Wild-Type Cutaneous Melanoma. Am J Clin Dermatol 2024; 25:407-419. [PMID: 38329690 DOI: 10.1007/s40257-023-00841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 02/09/2024]
Abstract
There have been many recent advances in melanoma therapy. While 50% of melanomas have a BRAF mutation and are a target for BRAF inhibitors, the remaining 50% are BRAF wild-type. Immune checkpoint inhibitors targeting PD-1, cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and lymphocyte activated gene-3 (Lag-3) are all approved for the treatment of patients with advanced BRAF wild-type melanoma; however, treatment of this patient population following initial immune checkpoint blockade is a current therapeutic challenge given the lack of other efficacious options. Here, we briefly review available US FDA-approved therapies for BRAF wild-type melanoma and focus on developing treatment avenues for this heterogeneous group of patients. We review the basics of genomic features of both BRAF mutant and BRAF wild-type melanoma as well as efforts underway to develop new targeted therapies involving the mitogen-activated protein kinase (MAPK) pathway for patients with BRAF wild-type tumors. We then focus on novel immunotherapies, including developing checkpoint inhibitors and agonists, cytokine therapies, oncolytic viruses and tumor-infiltrating lymphocytes, all of which represent potential therapeutic avenues for patients with BRAF wild-type melanoma who progress on currently approved immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Alexandra Haugh
- Department of Medicine, University of California San Francisco, 550 16th Street, 6809, San Francisco, CA, 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Adil I Daud
- Department of Medicine, University of California San Francisco, 550 16th Street, 6809, San Francisco, CA, 94158, USA.
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Lin J, Lin ZQ, Zheng SC, Chen Y. Immune checkpoint inhibitor-associated gastritis: Patterns and management. World J Gastroenterol 2024; 30:1941-1948. [PMID: 38681126 PMCID: PMC11045486 DOI: 10.3748/wjg.v30.i14.1941] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/23/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are widely used due to their effectiveness in treating various tumors. Immune-related adverse events (irAEs) are defined as adverse effects resulting from ICI treatment. Gastrointestinal irAEs are a common type of irAEs characterized by intestinal side effects, such as diarrhea and colitis, which may lead to the cessation of ICIs. Although irAE gastritis is rarely reported, it may lead to serious complications such as gastrorrhagia. Furthermore, irAE gastritis is often difficult to identify early due to its diverse symptoms. Although steroid hormones and immunosuppressants are commonly used to reverse irAEs, the best regimen and dosage for irAE gastritis remains uncertain. In addition, the risk of recurrence of irAE gastritis after the reuse of ICIs should be considered. In this editorial, strategies such as early identification, pathological diagnosis, management interventions, and immunotherapy rechallenge are discussed to enable clinicians to better manage irAE gastritis and improve the prognosis of these patients.
Collapse
Affiliation(s)
- Jing Lin
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350000, Fujian Province, China
| | - Zhong-Qiao Lin
- Phase I Clinical Trial Ward, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350000, Fujian Province, China
| | - Shi-Cheng Zheng
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350000, Fujian Province, China
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350000, Fujian Province, China
| |
Collapse
|
22
|
Mao YT, Wang Y, Chen XX, Liu CJ, Bao Q. Comparative severe dermatologic toxicities of immune checkpoint inhibitors in malignant melanoma: A systematic review and network meta-analysis. J Cosmet Dermatol 2024; 23:1165-1177. [PMID: 38013634 DOI: 10.1111/jocd.16105] [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: 07/14/2023] [Revised: 10/22/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have advanced the therapeutic landscape for malignant melanoma patients. However, they can cause permanent and irreversible dermatologic immune-related adverse events (irAEs) that may lead to interruption of ICI treatment or become life-threatening. To assess the risk of severe dermatologic irAEs (grade 3 or higher) among ICIs for advanced melanoma, we conducted a network meta-analysis (NMA). METHODS Phase II/III randomized controlled clinical trials (RCTs) involving ICIs were retrieved from various databases, including PubMed, Embase, Cochrane Library, and Web of Science. These trials were published from the inception of databases to October 15, 2022. In addition, the risk of severe dermatologic irAEs associated with ICI types and doses was evaluated and compared by NMA. RESULTS This study included 20 Phase II/III RCTs with a total of 10 575 patients. The results indicated that ICIs carry a higher risk of severe dermatologic irAEs compared to chemotherapy. Additionally, the combinational therapy of Nivolumab + Ipilimumab was associated with a higher risk than ICI monotherapy. Comparatively, the latest treatment option involving dual ICI therapy with Relatlimab + Nivolumab showed a lower toxicity risk, but higher than Ipilimumab alone. Lastly, Nivolumab, at a dose of 3 mg/kg every 2 weeks, was observed as the lowest-risk dosing regimen for severe dermatologic irAEs in patients with advanced melanoma. CONCLUSION The findings suggest that Nivolumab (1 mg/kg) + Ipilimumab (3 mg/kg) administered every 3 weeks should be used cautiously in patients with advanced melanoma at high risk for dermatologic irAEs. While we recommend the preferred regimen of Nivolumab (dose = 3 mg/kg, every 2 weeks).
Collapse
Affiliation(s)
- Yun-Tao Mao
- Department of Nursing, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Wang
- Department of Nursing, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Xiao Chen
- Department of Nursing, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng-Jiang Liu
- Department of General Medicine, Affiliated Anqing First People's Hospital of Anhui Medical University, Anqing, China
| | - Qi Bao
- Department of Plastic and Reconstructive Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
23
|
Riveiro-Barciela M, Carballal S, Díaz-González Á, Mañosa M, Gallego-Plazas J, Cubiella J, Jiménez-Fonseca P, Varela M, Menchén L, Sangro B, Fernández-Montes A, Mesonero F, Rodríguez-Gandía MÁ, Rivera F, Londoño MC. Management of liver and gastrointestinal toxicity induced by immune checkpoint inhibitors: Position statement of the AEEH-AEG-SEPD-SEOM-GETECCU. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:401-432. [PMID: 38228461 DOI: 10.1016/j.gastrohep.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/28/2023] [Accepted: 10/19/2023] [Indexed: 01/18/2024]
Abstract
The development of the immune checkpoint inhibitors (ICI) is one of the most remarkable achievements in cancer therapy in recent years. However, their exponential use has led to an increase in immune-related adverse events (irAEs). Gastrointestinal and liver events encompass hepatitis, colitis and upper digestive tract symptoms accounting for the most common irAEs, with incidence rates varying from 2% to 40%, the latter in patients undergoing combined ICIs therapy. Based on the current scientific evidence derived from both randomized clinical trials and real-world studies, this statement document provides recommendations on the diagnosis, treatment and prognosis of the gastrointestinal and hepatic ICI-induced adverse events.
Collapse
Affiliation(s)
- Mar Riveiro-Barciela
- Liver Unit, Internal Medicine Department, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Universitat Autònoma de Barcelona (UAB), Department of Medicine, Spain.
| | - Sabela Carballal
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Gastroenterology Department, Hospital Clinic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Spain; Universitat de Barcelona, Spain
| | - Álvaro Díaz-González
- Gastroenterology Department, Grupo de Investigación Clínica y Traslacional en Enfermedades Digestivas, Instituto de Investigación Valdecilla (IDIVAL), Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Míriam Mañosa
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Gastroenterology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | - Joaquín Cubiella
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Gastroenterology Department, Hospital Universitario de Ourense, Grupo de Investigación en Oncología Digestiva-Ourense, Spain
| | - Paula Jiménez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - María Varela
- Gastroenterology Department, Hospital Universitario Central de Asturias, IUOPA, ISPA, FINBA, University of Oviedo, Oviedo, Spain
| | - Luis Menchén
- Servicio de Aparato Digestivo - CEIMI, Instituto de Investigación Sanitaria Gregorio, Marañón, Spain; Departamento de Medicina, Universidad Complutense, Madrid, Spain
| | - Bruno Sangro
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Liver Unit, Cancer Center Clinica Universidad de Navarra, Pamplona-Madrid, Spain
| | - Ana Fernández-Montes
- Medical Oncology Department, Complexo Hospitalario Universitario de Ourense, Ourense, Spain
| | - Francisco Mesonero
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain; Universidad de Alcalá de Henares, Spain
| | - Miguel Ángel Rodríguez-Gandía
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), Madrid, Spain
| | - Fernando Rivera
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - María-Carlota Londoño
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Universitat de Barcelona, Spain; Liver Unit, Hospital Clínic Barcelona, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Spain
| |
Collapse
|
24
|
Ninmer EK, Zhu H, Chianese-Bullock KA, von Mehren M, Haas NB, Ross MI, Dengel LT, Slingluff CL. Multipeptide vaccines for melanoma in the adjuvant setting: long-term survival outcomes and post-hoc analysis of a randomized phase II trial. Nat Commun 2024; 15:2570. [PMID: 38519525 PMCID: PMC10959948 DOI: 10.1038/s41467-024-46877-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024] Open
Abstract
The critical roles of CD4+ T cells have been understudied for cancer vaccines. Here we report long-term clinical outcomes of a randomized multicenter phase II clinical trial (NCT00118274), where patients with high-risk melanoma received a multipeptide vaccine targeting CD8+ T cells (12MP) and were randomized to receive either of two vaccines for CD4+ (helper) T cells: 6MHP (6 melanoma-specific helper peptides), or tet (a nonspecific helper peptide from tetanus toxoid). Cyclophosphamide (Cy) pre-treatment was also assessed. Primary outcomes for T cell responses to 12MP, 6MHP, and tet were previously reported, suggesting immunogenicity of both vaccines but that CD8 T cell responses to 12MP were lower when tet was replaced with 6MHP. Here, in post-hoc analyses, we report durable prolongation of overall survival by adding 6MHP instead of tet. That benefit was experienced only by male patients. A favorable interaction of 6MHP and Cy is also suggested. Multivariable Cox regression analysis of the intent-to-treat population identify vaccine arm (12MP + 6MHP+Cy) and patient sex (male) as the two significant predictors of enhanced survival. These findings support the value of adding cognate T cell help to cancer vaccines and also suggest a need to assess the impact of patient sex on immune therapy outcomes.
Collapse
Affiliation(s)
- Emily K Ninmer
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Hong Zhu
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- University of Virginia, School of Medicine, Cancer Center, Charlottesville, VA, USA
| | - Kimberly A Chianese-Bullock
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA
- University of Virginia, School of Medicine, Cancer Center, Charlottesville, VA, USA
| | | | - Naomi B Haas
- Fox Chase Cancer Center, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Merrick I Ross
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Lynn T Dengel
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Craig L Slingluff
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA.
- University of Virginia, School of Medicine, Cancer Center, Charlottesville, VA, USA.
| |
Collapse
|
25
|
Ayass MA, Tripathi T, Griko N, Okyay T, Ramankutty Nair R, Zhang J, Zhu K, Melendez K, Pashkov V, Abi-Mosleh L. Dual Checkpoint Aptamer Immunotherapy: Unveiling Tailored Cancer Treatment Targeting CTLA-4 and NKG2A. Cancers (Basel) 2024; 16:1041. [PMID: 38473398 DOI: 10.3390/cancers16051041] [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: 01/23/2024] [Revised: 02/13/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Recent strides in immunotherapy have illuminated the crucial role of CTLA-4 and PD-1/PD-L1 pathways in contemporary oncology, presenting both promises and challenges in response rates and adverse effects. This study employs a computational biology tool (in silico approach) to craft aptamers capable of binding to dual receptors, namely, inhibitory CTLA4 and NKG2A, thereby unleashing both T and NK cells and enhancing CD8+ T and NK cell functions for tumor cell lysis. Computational analysis highlighted AYA22T-R2-13 with HADDOCK scores of -78.2 ± 10.2 (with CTLA4), -60.0 ± 4.2 (with NKG2A), and -77.5 ± 5.6 (with CD94/NKG2A). Confirmation of aptamer binding to targeted proteins was attained via ELISA and flow cytometry methods. In vitro biological functionality was assessed using lactate dehydrogenase (LDH) cytotoxicity assay. Direct and competitive assays using ELISA and flow cytometry demonstrated the selective binding of AYA22T-R2-13 to CTLA4 and NKG2A proteins, as well as to the cell surface receptors of IL-2-stimulated T cells and NK cells. This binding was inhibited in the presence of competition from CTLA4 or NKG2A proteins. Remarkably, the blockade of CTLA4 or NKG2A by AYA22T-R2-13 augmented human CD8 T cell- and NK cell-mediated tumor cell lysis in vitro. Our findings highlight the precise binding specificity of AYA22T-R2-13 for CTLA4-B7-1/B7-2 (CD80/CD86) or CD94/NKG2A-HLA-E interactions, positioning it as a valuable tool for immune checkpoint blockade aptamer research in murine tumor models. These in vitro studies establish a promising foundation for further enhancing binding capacity and establishing efficacy and safety in animal models. Consequently, our results underscore the potential of AYA22T-R2-13 in cancer immunotherapy, offering high specificity, low toxicity, and the potential for cost-effective production.
Collapse
Affiliation(s)
| | | | - Natalya Griko
- Ayass Bioscience LLC, 8501 Wade Blvd, Bld 9, Frisco, TX 75034, USA
| | - Tutku Okyay
- Ayass Bioscience LLC, 8501 Wade Blvd, Bld 9, Frisco, TX 75034, USA
| | | | - Jin Zhang
- Ayass Bioscience LLC, 8501 Wade Blvd, Bld 9, Frisco, TX 75034, USA
| | - Kevin Zhu
- Ayass Bioscience LLC, 8501 Wade Blvd, Bld 9, Frisco, TX 75034, USA
| | - Kristen Melendez
- Ayass Bioscience LLC, 8501 Wade Blvd, Bld 9, Frisco, TX 75034, USA
| | - Victor Pashkov
- Ayass Bioscience LLC, 8501 Wade Blvd, Bld 9, Frisco, TX 75034, USA
| | - Lina Abi-Mosleh
- Ayass Bioscience LLC, 8501 Wade Blvd, Bld 9, Frisco, TX 75034, USA
| |
Collapse
|
26
|
Ku PY, Hsieh PY, Wu FH. Gallbladder metastases from primary anorectal malignant melanoma: A case report. Asian J Surg 2024; 47:1538-1539. [PMID: 38065737 DOI: 10.1016/j.asjsur.2023.11.165] [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: 10/31/2023] [Accepted: 11/24/2023] [Indexed: 03/13/2024] Open
Affiliation(s)
- Peng-Yu Ku
- Division of General Surgery, Department of Surgery, Taichung Veterans General Hospital, No. 1650, Sec. 4, Taiwan Boulevard, Situn District, Taichung City, 407219, Taiwan, ROC
| | - Po-Yen Hsieh
- Division of General Surgery, Department of Surgery, Taichung Veterans General Hospital, No. 1650, Sec. 4, Taiwan Boulevard, Situn District, Taichung City, 407219, Taiwan, ROC; Department of Urology, Taichung Veterans General Hospital, No. 1650, Sec. 4, Taiwan Boulevard, Situn District, Taichung City, 407219, Taiwan, ROC; School of Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei City, Taiwan, 110, Taiwan, ROC
| | - Feng-Hsu Wu
- Division of General Surgery, Department of Surgery, Taichung Veterans General Hospital, No. 1650, Sec. 4, Taiwan Boulevard, Situn District, Taichung City, 407219, Taiwan, ROC; School of Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei City, Taiwan, 110, Taiwan, ROC.
| |
Collapse
|
27
|
Zhou P, Gao Y, Kong Z, Wang J, Si S, Han W, Li J, Lv Z, Wang R. Immune checkpoint inhibitors and acute kidney injury. Front Immunol 2024; 15:1353339. [PMID: 38464524 PMCID: PMC10920224 DOI: 10.3389/fimmu.2024.1353339] [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: 12/10/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024] Open
Abstract
As a new type of anti-tumor immunotherapy, immune checkpoint inhibitors (ICIs) have improved the prognosis of multiple malignancies. However, renal complications are becoming more frequent. Nephrotoxicity often manifests as acute kidney injury (AKI), and the most common histopathological type is acute tubulointerstitial nephritis (ATIN). Based on previous studies of the incidence and potential risk factors for nephrotoxicity, in this review, we describe the mechanism of AKI after ICIs treatment, summarize the incidence, risk factors, and outcomes of AKI, and discuss the diagnosis and management of immune checkpoint inhibitors-associated acute kidney injury (ICI-AKI). In addition, we review the current status of ICIs rechallenge and the therapeutic strategies of ICIs applied in kidney transplant recipients. Finally, we emphasize the importance of collaboration between nephrologists and oncologists to guide the treatment of ICIs and the management of renal complications.
Collapse
Affiliation(s)
- Ping Zhou
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Ying Gao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhijuan Kong
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Junlin Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuxuan Si
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wei Han
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Li
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
28
|
Luo X, Zhang N, Guo L, Zhou L, Jiang H, Cui RS. Comprehensive needs, social support, and disease perception in lung cancer patients treated with immune checkpoint inhibitors: a cross-sectional study. Support Care Cancer 2024; 32:166. [PMID: 38372773 DOI: 10.1007/s00520-024-08348-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/30/2024] [Indexed: 02/20/2024]
Abstract
PURPOSE The aim of this study was to investigate the comprehensive needs of lung cancer patients treated with immune checkpoint inhibitors and to explore the relationships between comprehensive needs and social support and disease perception, moreover, to analyse associated factors of comprehensive needs. METHODS The study was conducted in a teaching hospital in Jiaxing Province, China. A total of 141 patients with lung cancer completed a battery of self-report questionnaires, including the Comprehensive Needs Assessment Tool in Cancer for Patients (CNAT), Social Supportive Rating Scale (SSRS), Brief Illness Perception Questionnaire (BIPQ), and demographic and clinical characteristics questionnaire. RESULTS The level of comprehensive needs was highest in the domain "medical demand" (42.17 ± 26.57), and the item with the highest level of comprehensive needs was "I need information about the financial support for my medical expenses" (2.00 ± 1.07). Statistically significant correlations were identified between the comprehensive needs score, social support, and disease perception. The multiple regression analysis showed that immunotherapy course, whether irAEs occur, social support, and disease perception were factors influencing patients' comprehensive needs. CONCLUSIONS The most prevalent needs in lung cancer patients were found in the "medical needs" domain. Additionally, immunotherapy course, whether irAEs occur, disease perception, and social support were associated with comprehensive needs among lung cancer patients. It is essential to combine the associated factors to accurately evaluate patient needs. We should pay more attention to proposing the comprehensive measures for these patients and providing more individualized supportive care during the lengthy treatment period.
Collapse
Affiliation(s)
- Xiaoyan Luo
- Department of Nursing, Medical College, Jiaxing College, No.899, Guangdome Road, Nanhu District, Jiaxing, Zhejiang Province, China
| | - Ningning Zhang
- Department of Medical Oncology, The First Hospital of Jiaxing, South Zhonghuan Road, Jiaxing, 1882314001, Zhejiang Province, China
| | - Lingru Guo
- Department of Nursing, Medical College, Jiaxing College, No.899, Guangdome Road, Nanhu District, Jiaxing, Zhejiang Province, China
| | - Li Zhou
- Department of Nursing, Medical College, Jiaxing College, No.899, Guangdome Road, Nanhu District, Jiaxing, Zhejiang Province, China
| | - Haiying Jiang
- Department of Nursing, Medical College, Jiaxing College, No.899, Guangdome Road, Nanhu District, Jiaxing, Zhejiang Province, China
| | - Ren-Shan Cui
- Department of Nursing, Medical College, Jiaxing College, No.899, Guangdome Road, Nanhu District, Jiaxing, Zhejiang Province, China.
| |
Collapse
|
29
|
Lasagna A, Sacchi P. The ABC of Immune-Mediated Hepatitis during Immunotherapy in Patients with Cancer: From Pathogenesis to Multidisciplinary Management. Cancers (Basel) 2024; 16:795. [PMID: 38398187 PMCID: PMC10886483 DOI: 10.3390/cancers16040795] [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: 11/16/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Immune-mediated hepatotoxicity (IMH) is not-so-rare complication during treatment with immune checkpoint inhibitors (ICIs). This narrative review aims to report the current knowledge on hepatic immune-related adverse events (irAEs) during immunotherapy from pathogenesis to multidisciplinary management. The majority of cases of IMH are asymptomatic and only a few patients may have clinical conditions. The severity of IMH is usually stratified according to Common Terminology for Clinical Adverse Events (CTCAE) criteria, but these scores may overestimate the clinical severity of IMH compared to the Drug-Induced Liver Injury Network (DILIN) scale. The differential diagnosis of IMH is challenging because the elevated liver enzymes can be due to a number of etiologies such as viral infection, autoimmune and metabolic diseases, liver metastases, biliary diseases, and other drugs. The cornerstones of IMH management are represented by withholding or delaying ICI administration and starting immunosuppressive therapy. A multidisciplinary team, including oncologists, hepatologists, internists, and emergency medicine physicians, is essential for the management of IMH.
Collapse
Affiliation(s)
- Angioletta Lasagna
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Paolo Sacchi
- Division of Infectious Diseases I, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
30
|
Riveiro-Barciela M, Carballal S, Díaz-González Á, Mañosa M, Gallgo-Plazas J, Cubiella J, Jiménez-Fonseca P, Varela M, Menchén L, Sangro B, Fernández-Montes A, Mesonero F, Rodríguez-Gandía MÁ, Rivera F, Londoño MC. Management of liver and gastrointestinal toxicity induced by immune checkpoint inhibitors: Position statement of the AEEH-AEG-SEPD-SEOM-GETECCU. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 116:83-113. [PMID: 38226597 DOI: 10.17235/reed.2024.10250/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The development of the immune checkpoint inhibitors (ICI) is one of the most remarkable achievements in cancer therapy in recent years. However, their exponential use has led to an increase in immune-related adverse events (irAEs). Gastrointestinal and liver events encompass hepatitis, colitis and upper digestive tract symptoms accounting for the most common irAEs, with incidence rates varying from 2 % to 40 %, the latter in patients undergoing combined ICIs therapy. Based on the current scientific evidence derived from both randomized clinical trials and real-world studies, this statement document provides recommendations on the diagnosis, treatment and prognosis of the gastrointestinal and hepatic ICI-induced adverse events.
Collapse
Affiliation(s)
| | | | | | - Miriam Mañosa
- Gastroenterology, Hospital Universitari Germans Trias i Pujol
| | | | | | | | - María Varela
- Gastroenterology, Hospital Universitario Central de Asturias
| | - Luis Menchén
- Digestive Diseases, Instituto de Investigación Sanitaria Gregorio Marañón
| | | | | | | | | | - Fernando Rivera
- Hospital Universitario Marqués de Valdecilla, Medical Oncology
| | | |
Collapse
|
31
|
Harvey C, Nahar KJ, McKeown J, Lo SN, Farag S, Yousaf N, Young K, Tas L, Meerveld-Eggink A, Blank C, Thomas A, McQuade J, Schilling B, Johnson DB, Huertas RM, Arance A, Lee J, Zimmer L, Long GV, Carlino MS, Wang Y, Menzies AM. Management of infliximab refractory immune checkpoint inhibitor gastrointestinal toxicity: a multicenter case series. J Immunother Cancer 2024; 12:e008232. [PMID: 38296594 PMCID: PMC10831444 DOI: 10.1136/jitc-2023-008232] [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: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) gastrointestinal toxicity (gastritis, enteritis, colitis) is a major cause of morbidity and treatment-related death. Guidelines agree steroid-refractory cases warrant infliximab, however best management of infliximab-refractory ICI gastrointestinal toxicity (IRIGItox) is unknown. METHODS We conducted an international multicenter retrospective case series. IRIGItox was defined as failure of symptom resolution ≤grade 1 (Common Terminology Criteria for Adverse Events V.5.0) following ≥2 infliximab doses or failure of symptom resolution ≤grade 2 after one dose. Data were extracted regarding demographics, steroid use, response to treatment, and survival outcomes. Toxicity was graded at symptom onset and time of infliximab failure. Efficacy of infliximab refractory therapy was assessed by symptom resolution, time to resolution and steroid wean duration. Survival outcomes were examined based on immunosuppressive therapy received. RESULTS 78 patients were identified: median age 60 years; 56% men; majority melanoma (N=70, 90%); 60 (77%) received anti-cytotoxic T-lymphocyte-associated protein 4 alone or in combination with anti-programmed cell death protein-1 and most had colitis (N=74, 95%). 106 post-infliximab treatments were given: 31 calcineurin inhibitors (CNIs); 27 antimetabolites (mycophenolate, azathioprine); 16 non-systemic immunomodulatory agents (eg, mesalazine or budesonide); 15 vedolizumab; 5 other biologics (anti-interleukin-12/23, 16, Janus kinase inhibitors) and 7 interventional procedures (including colectomy); 5 did not receive post-infliximab therapy. Symptom resolution was achieved in most (N=23/31, 74%) patients treated with CNIs; 12/27 (44%) with antimetabolites; 7/16 (44%) with non-systemic immunomodulation, 8/15 (53%) with vedolizumab and 5/7 (71%) with interventional procedures. No non-vedolizumab biologics resulted in toxicity resolution. CNIs had the shortest time to symptom resolution (12 days) and steroid wean (43 days); however, were associated with poorer event-free survival (6.3 months) and overall survival (26.8 months) than other agents. Conversely, vedolizumab had the longest time to toxicity resolution and steroid wean, 66 and 124 days, but most favorable survival data: EFS 24.5 months; median OS not reached. Six death occurred (three due to IRIGItox or management of toxicity; three with persisting IRIGItox and progressive disease). CONCLUSIONS IRIGItox causes major morbidity and mortality. Management is heterogeneous. CNIs appear most likely to result in toxicity resolution in the shortest time period, however, are associated with poorer oncological outcomes in contrast to vedolizumab.
Collapse
Affiliation(s)
- Catriona Harvey
- Westmead Hospital WNH, Westmead, New South Wales, Australia
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
| | - Kazi J Nahar
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
- The University of Sydney, Sydney, New South Wales, Australia
| | - Janet McKeown
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
- The University of Sydney, Sydney, New South Wales, Australia
| | - Serigne N Lo
- Research and Biostatistics Group, Melanoma Institute Australia, North Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | | | | | - Kate Young
- Royal Marsden Hospital NHS Trust, London, UK
| | - Liselotte Tas
- Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Austin Thomas
- The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jennifer McQuade
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | - Ana Arance
- Hospital Clinic and Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Joanna Lee
- The University of Sydney, Sydney, New South Wales, Australia
- Westmead Hospital, Westmead, New South Wales, Australia
| | - Lisa Zimmer
- Dermatology, University Hospital Essen, Essen, Germany
| | - Georgina V Long
- Melanoma Institute Australia, Wollstonecraft, New South Wales, Australia
| | | | - Yinghong Wang
- Gastroenterology, Hepatology & Nutrition, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alexander Maxwell Menzies
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
- The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
32
|
Curkovic NB, Bai K, Ye F, Johnson DB. Incidence of Cutaneous Immune-Related Adverse Events and Outcomes in Immune Checkpoint Inhibitor-Containing Regimens: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:340. [PMID: 38254829 PMCID: PMC10814132 DOI: 10.3390/cancers16020340] [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: 11/06/2023] [Revised: 12/19/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are used to treat many cancers, and cutaneous immune-related adverse events (cirAEs) are among the most frequently encountered toxic effects. Understanding the incidence and prognostic associations of cirAEs is of importance as their uses in different settings, combinations, and tumor types expand. To evaluate the incidence of cirAEs and their association with outcome measures across a variety of ICI regimens and cancers, we performed a systematic review and meta-analysis of published trials of anti-programmed death-1/ligand-1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) ICIs, both alone and in combination with chemotherapy, antiangiogenic agents, or other ICIs in patients with melanoma, renal cell carcinoma, non-small cell lung cancer, and urothelial carcinoma. Key findings of our study include variable cirAE incidence among tumors and ICI regimens, positive association with increased cirAE incidence and response rate, as well as significant association between increased vitiligo incidence and overall survival. Across 174 studies, rash, pruritis, and vitiligo were the most reported cirAEs, with incidences of 16.7%, 18.0%, and 6.6%, respectively. Higher incidence of cirAEs was associated with ICI combination regimens and with CTLA-4-containing regimens, particularly with higher doses of ipilimumab, as compared to PD-1/L1 monotherapies. Outcome measures including response rate and progression-free survival were positively correlated with incidence of cirAEs. The response rate and incidence of pruritis, vitiligo, and rash were associated with expected rises in incidence of 0.17% (p = 0.0238), 0.40% (p = 0.0010), and 0.18% (p = 0.0413), respectively. Overall survival was positively correlated with the incidence of pruritis, vitiligo, and rash; this association was significant for vitiligo (p = 0.0483). Our analysis provides benchmark incidence rates for cirAEs and links cirAEs with favorable treatment outcomes at a study level across diverse solid tumors and multiple ICI regimens.
Collapse
Affiliation(s)
- Nina B. Curkovic
- School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Kun Bai
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Fei Ye
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Douglas B. Johnson
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
33
|
Su J, Fu Y, Cui Z, Abidin Z, Yuan J, Zhang X, Li R, Zhao C. Relatlimab: a novel drug targeting immune checkpoint LAG-3 in melanoma therapy. Front Pharmacol 2024; 14:1349081. [PMID: 38269271 PMCID: PMC10806167 DOI: 10.3389/fphar.2023.1349081] [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: 12/04/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Relatlimab is a type of human immunoglobulin G4 monoclonal blocking antibody. It is the world's first Lymphocyte-Activation Gene-3 (LAG-3) inhibitor and the third immune checkpoint inhibitor with clinical application, following PD-1 and CTLA-4. Relatlimab can bind to the LAG-3 receptor which blocks the interaction between LAG-3 and its ligand to reduce LAG-3 pathway-mediated immunosuppression and promote T-cell proliferation, inducing tumor cell death. On 18 March 2022, the U.S. FDA approved the fixed-dose combination of relatlimab developed by Bristol Myers Squibb with nivolumab, under the brand name Opdualag for the treatment of unresectable or metastatic melanoma in adult and pediatric patients aged 12 and older. This study comprehensively describes the mechanism of action and clinical trials of relatlimab and a brief overview of immune checkpoint drugs currently used for the treatment of melanoma.
Collapse
Affiliation(s)
- Jingjing Su
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Yiting Fu
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zitong Cui
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zain Abidin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jingsong Yuan
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Xinmiao Zhang
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Runmin Li
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Chunzhen Zhao
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
34
|
Amin T, Hossain A, Jerin N, Mahmud I, Rahman MA, Rafiqul Islam SM, Islam SMBUL. Immunoediting Dynamics in Glioblastoma: Implications for Immunotherapy Approaches. Cancer Control 2024; 31:10732748241290067. [PMID: 39353594 PMCID: PMC11459535 DOI: 10.1177/10732748241290067] [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: 02/20/2024] [Revised: 09/14/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Glioblastoma is an aggressive primary brain tumor that poses many therapeutic difficulties because of the high rate of proliferation, genetic variability, and its immunosuppressive microenvironment. The theory of cancer immunoediting, which includes the phases of elimination, equilibrium, and escape, offers a paradigm for comprehending interactions between the immune system and glioblastoma. Immunoediting indicates the process by which immune cells initially suppress tumor development, but thereafter select for immune-resistant versions leading to tumor escape and progression. The tumor microenvironment (TME) in glioblastoma is particularly immunosuppressive, with regulatory T cells and myeloid-derived suppressor cells being involved in immune escape. To achieve an efficient immunotherapy for glioblastoma, it is crucial to understand these mechanisms within the TME. Existing immunotherapeutic modalities such as chimeric antigen receptor T cells and immune checkpoint inhibitors have been met with some level of resistance because of the heterogeneous nature of the immune response to glioblastoma. Solving these issues is critical to develop novel strategies capable of modulating the TME and re-establishing normal immune monitoring. Further studies should be conducted to identify the molecular and cellular events that underlie the immunosuppressive tumor microenvironment in glioblastoma. Comprehending and modifying the stages of immunoediting in glioblastoma could facilitate the development of more potent and long-lasting therapies.
Collapse
Affiliation(s)
- Tasbir Amin
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Amana Hossain
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Nusrat Jerin
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Imteaz Mahmud
- Department of Public Health, North South University, Dhaka, Bangladesh
| | - Md Ahasanur Rahman
- Department of Physiology and Biophysics, Howard University, College of Medicine, Washington, DC, USA
| | - SM Rafiqul Islam
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda, USA
| | - S M Bakhtiar UL Islam
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| |
Collapse
|
35
|
Stockem CF, Galsky MD, van der Heijden MS. Turning up the heat: CTLA4 blockade in urothelial cancer. Nat Rev Urol 2024; 21:22-34. [PMID: 37608154 DOI: 10.1038/s41585-023-00801-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/24/2023]
Abstract
Anti-PD1 and anti-PDL1 monotherapies have shown clinical efficacy in stage IV urothelial cancer and are integrated into current clinical practice. However, only a small number of the patients treated with single-agent checkpoint blockade experience an antitumour response. Insufficient priming or inhibitory factors in the tumour immune microenvironment might have a role in the lack of response. CTLA4 is an inhibitory checkpoint on activated T cells that is being studied as a therapeutic target in combination with anti-PD1 or anti-PDL1 therapies in advanced urothelial cancer. In locally advanced urothelial cancer, this combination approach has shown encouraging antitumour effects when administered pre-operatively. We believe that the presence of pre-existing intratumoural T cell immunity is not a prerequisite for response to combination therapy and that the additional value of CTLA4 blockade might involve the broadening of peripheral T cell priming, thereby transforming immunologically cold tumours into hot tumours.
Collapse
Affiliation(s)
- Chantal F Stockem
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Matthew D Galsky
- Department of Genitourinary Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | | |
Collapse
|
36
|
Kejamurthy P, Devi KTR. Immune checkpoint inhibitors and cancer immunotherapy by aptamers: an overview. Med Oncol 2023; 41:40. [PMID: 38158454 DOI: 10.1007/s12032-023-02267-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024]
Abstract
Efforts in cancer immunotherapy aim to counteract evasion mechanisms and stimulate the immune system to recognise and attack cancer cells effectively. Combination therapies that target multiple aspects of immune evasion are being investigated to enhance the overall efficacy of cancer immunotherapy. PD-1 (Programmed Cell Death Protein 1), CTLA-4 (Cytotoxic T-Lymphocyte Antigen 4), LAG-3 (Lymphocyte-Activation Gene 3), and TIM-3 (T Cell Immunoglobulin and Mucin Domain-Containing Protein3) are all immune checkpoint receptors that play crucial roles in regulating the immune response and maintaining self-tolerance often exploited by cancer cells to evade immune surveillance. Antibodies targeted against immune checkpoint inhibitors such as anti-PD-1 antibodies (e.g., pembrolizumab, nivolumab), anti-CTLA-4 antibodies (e.g., Ipilimumab), and experimental drugs targeting LAG-3 and TIM-3, aim to block these interactions and unleash the immune system's ability to recognise and destroy cancer cells. The US FDA has approved different categories of immune checkpoint inhibitors that have been utilised successfully in some patients with metastatic melanoma, renal cell carcinoma, head and neck cancers, and non-small lung cancer. Although several immune checkpoint inhibitor antibodies have been developed, they exhibited immune-related adverse effects, resulting in hypophysitis, diabetes, and neurological issues. These adverse effects of antibodies can be reduced by developing aptamer against the target. Aptamers offer several advantages over traditional antibodies, such as improved specificity, reduced immunogenicity, and flexible design for reduced adverse effects that specifically target and block protein-protein or receptor-ligand interactions involved in immune checkpoint pathways. The current study aims to review the function of particular immune checkpoint inhibitors along with developed aptamer-mediated antitumor cytotoxicity in cancer treatment.
Collapse
Affiliation(s)
- Priyatharcini Kejamurthy
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - K T Ramya Devi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| |
Collapse
|
37
|
Jenkins KA, Park M, Pederzoli-Ribeil M, Eskiocak U, Johnson P, Guzman W, McLaughlin M, Moore-Lai D, O'Toole C, Liu Z, Nicholson B, Flesch V, Qiu H, Clackson T, O'Hagan RC, Rodeck U, Karow M, O'Neil J, Williams JC. XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancer. J Immunother Cancer 2023; 11:e007785. [PMID: 38164757 PMCID: PMC10729150 DOI: 10.1136/jitc-2023-007785] [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: 11/09/2023] [Indexed: 01/03/2024] Open
Abstract
INTRODUCTION The clinical benefit of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab has been well established but limited by immune-related adverse events, especially when ipilimumab is used in combination with anti-PD-(L)1 mAb therapy. To overcome these limitations, we have developed XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 mAb. METHODS XTX101 consists of an anti-human CTLA-4 mAb covalently linked to masking peptides that block the complementarity-determining regions, thereby minimizing the mAb binding to CTLA-4. The masking peptides are designed to be released by proteases that are typically dysregulated within the tumor microenvironment (TME), resulting in activation of XTX101 intratumorally. Mutations within the Fc region of XTX101 were included to enhance affinity for FcγRIII, which is expected to enhance potency through antibody-dependent cellular cytotoxicity. RESULTS Biophysical, biochemical, and cell-based assays demonstrate that the function of XTX101 depends on proteolytic activation. In human CTLA-4 transgenic mice, XTX101 monotherapy demonstrated significant tumor growth inhibition (TGI) including complete responses, increased intratumoral CD8+T cells, and regulatory T cell depletion within the TME while maintaining minimal pharmacodynamic effects in the periphery. XTX101 in combination with anti-PD-1 mAb treatment resulted in significant TGI and was well tolerated in mice. XTX101 was activated in primary human tumors across a range of tumor types including melanoma, renal cell carcinoma, colon cancer and lung cancer in an ex vivo assay system. CONCLUSIONS These data demonstrate that XTX101 retains the full potency of an Fc-enhanced CTLA-4 antagonist within the TME while minimizing the activity in non-tumor tissue, supporting the further evaluation of XTX101 in clinical studies.
Collapse
Affiliation(s)
- Kurt A Jenkins
- Xilio Therapeutics, Waltham, Massachusetts, USA
- Molecular Medicine, City of Hope National Medical Center, Beckman Research Institute, Duarte, California, USA
| | - Miso Park
- Molecular Medicine, City of Hope National Medical Center, Beckman Research Institute, Duarte, California, USA
| | | | | | - Parker Johnson
- Xilio Therapeutics, Waltham, Massachusetts, USA
- Molecular Medicine, City of Hope National Medical Center, Beckman Research Institute, Duarte, California, USA
| | | | | | | | | | - Zhen Liu
- Xilio Therapeutics, Waltham, Massachusetts, USA
| | | | - Veronica Flesch
- Molecular Medicine, City of Hope National Medical Center, Beckman Research Institute, Duarte, California, USA
| | - Huawei Qiu
- Xilio Therapeutics, Waltham, Massachusetts, USA
| | | | | | - Ulrich Rodeck
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | - John C Williams
- Molecular Medicine, City of Hope National Medical Center, Beckman Research Institute, Duarte, California, USA
| |
Collapse
|
38
|
Niessner H, Hüsch A, Kosnopfel C, Meinhardt M, Westphal D, Meier F, Schilling B, Sinnberg T. Exploring the In Vitro and In Vivo Therapeutic Potential of BRAF and MEK Inhibitor Combination in NRAS-Mutated Melanoma. Cancers (Basel) 2023; 15:5521. [PMID: 38067230 PMCID: PMC10705743 DOI: 10.3390/cancers15235521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 10/16/2024] Open
Abstract
INTRODUCTION Patients with NRAS-mutant metastatic melanoma often have an aggressive disease requiring a fast-acting, effective therapy. The MEK inhibitor binimetinib shows an overall response rate of 15% in patients with NRAS-mutant melanoma, providing a backbone for combination strategies. Our previous studies demonstrated that in NRAS-mutant melanoma, the antitumor activity of the MEK inhibitor binimetinib was significantly potentiated by the BRAFV600E/K inhibitor encorafenib through the induction of ER stress, leading to melanoma cell death by apoptotic mechanisms. Encorafenib combined with binimetinib was well tolerated in a phase III trial showing potent antitumor activity in BRAF-mutant melanoma, making a rapid evaluation in NRAS-mutant melanoma imminently feasible. These data provide a mechanistic rationale for the evaluation of binimetinib combined with encorafenib in preclinical and clinical studies on NRAS-mutant metastatic melanoma. METHODS The combination of BRAFi plus MEKi was tested in a monolayer culture of patient-derived cell lines and in corresponding patient-derived tissue slice cultures of NRAS-mutant melanoma. To investigate the treatment in vivo, NSG (NOD. Cg-PrkdcscidIl2rgtm1Wjl/SzJ) mice were subcutaneously injected with three different BRAF wild-type melanoma models harboring oncogenic NRAS mutations and treated orally with encorafenib (6 mg/kg body weight, daily) with or without binimetinib (8 mg/kg body weight, twice daily). In parallel, an individual healing attempt was carried out by treating one patient with an NRAS-mutated tumor. RESULTS Encorafenib was able to enhance the inhibitory effect on cell growth of binimetinib only in the cell line SKMel147 in vitro. It failed to enhance the apoptotic effect found in two other NRAS-mutated cell lines. Encorafenib led to a hyperactivation of ERK which could be reduced with the combinational treatment. In two of the three patient-derived tissue slice culture models of NRAS-mutant melanomas, a slight tendency of a combinatorial effect was seen which was not significant. Encorafenib showed a slight induction of the ER stress genes ATF4, CHOP, and NUPR1. The combinational treatment was able to enhance this effect, but not significantly. In the mouse model, the combination therapy of encorafenib with binimetinib resulted in reduced tumor growth compared to the control and encorafenib groups; however, the best effect in terms of tumor growth inhibition was measured in the binimetinib therapy group. The therapy showed no effect in an individual healing attempt for a patient suffering from metastatic, therapy-refractory NRAS-mutated melanoma. CONCLUSION In in vitro and ex vivo settings, the combination therapy was observed to elicit a response; however, it did not amplify the efficacy observed with binimetinib alone, whereas in a patient, the combinational treatment remained ineffective. The preclinical in vivo data showed no increased combinatorial effect. However, the in vivo effect of binimetinib as monotherapy was unexpectedly high in the tested regimen. Nevertheless, binimetinib proved to be advantageous in the treatment of melanoma in vivo and led to high rates of apoptosis in vitro; hence, it still seems to be a good base for combination with other substances in the treatment of patients with NRAS-mutant melanoma.
Collapse
Affiliation(s)
- Heike Niessner
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
| | - Anna Hüsch
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
| | - Corinna Kosnopfel
- Department of Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany;
| | - Matthias Meinhardt
- Department of Pathology, Medical Faculty and University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany;
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
| | - Dana Westphal
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, 01307 Dresden, Germany
| | - Friedegund Meier
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, 01307 Dresden, Germany
- Center for Regenerative Therapies Dresden, TU Dresden, 01307 Dresden, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany;
| | - Tobias Sinnberg
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
39
|
Gudd CLC, Sheth R, Thursz MR, Triantafyllou E, Possamai LA. Immune Checkpoint Inhibitor-Induced Liver Injury. Semin Liver Dis 2023; 43:402-417. [PMID: 38101418 DOI: 10.1055/s-0043-1776761] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
In recent years cancer treatment has been revolutionized by the development and wide application of checkpoint inhibitor (CPI) drugs, which are a form of immunotherapy. CPI treatment is associated with immune-related adverse events, off-target tissue destructive inflammatory complications, which may affect a range of organs, with liver inflammation (hepatitis) being one of the more commonly noted events. This is a novel form of drug-induced liver injury and a rapidly evolving field, as our understanding of both the basic immunopathology of CPI hepatitis (CPI-H) and optimal clinical management, races to catch up with the increasing application of this form of immunotherapy in clinical practice. In this review, we summarize current evidence and understanding of CPI-H, from fundamental immunology to practical patient management.
Collapse
Affiliation(s)
- Cathrin L C Gudd
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Roosey Sheth
- Institute of Liver Studies, Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Mark R Thursz
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Liver and Antiviral Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Lucia A Possamai
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Liver and Antiviral Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
40
|
Amoako-Teming P, Rostami P, Mehta P, Saeed I. Anorectal Melanoma: A Case Report. Cureus 2023; 15:e48835. [PMID: 38106750 PMCID: PMC10722867 DOI: 10.7759/cureus.48835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
Anorectal mucosal melanoma (AMM) is an infrequent and highly aggressive form of mucosal melanoma. Its rarity makes it challenging to clinically diagnose, and its initial symptoms are typically nonspecific such as rectal/anal bleeding (the most common symptom), anal pain, or the presence of an anal mass. The prognosis for this condition is generally poor, and its incidence appears to be increasing each year. AMMs often go undetected and/or are already metastasized at the time of diagnosis. We present a case report of a patient who initially presented with nonspecific symptoms of anemia and blood per rectum, and was later found to have stage IV melanoma of the anorectal region. There is a notable scarcity of literature on this disease, resulting in a lack of a comprehensive understanding of its nature. Most available information consists of isolated case reports rather than comprehensive studies. Although surgical resection remains the primary treatment approach, the majority of patients (over 80%) will die due to distant metastasis within five years after undergoing surgery. The five-year survival rate for anorectal melanoma is estimated to be between 6% and 22%.
Collapse
Affiliation(s)
- Papa Amoako-Teming
- Department of Surgery, St. Peter's University Hospital, New Brunswick, USA
- Department of Surgery, St. George's University School of Medicine, Great River, USA
| | - Pouya Rostami
- Department of Surgery, St. George's University School of Medicine, Great River, USA
- Department of Surgery, St. Peter's University Hospital, New Brunswick, USA
| | - Pawan Mehta
- Department of Surgery, St. George's University School of Medicine, Great River, USA
- Department of Surgery, St. Peter's University Hospital, New Brunswick, USA
| | - Imran Saeed
- Department of Surgery, St. Peter's University Hospital - Rutgers Robert Wood Johnson School of Medicine, New Brunswick, USA
| |
Collapse
|
41
|
Miao J, Herrmann SM. Immune checkpoint inhibitors and their interaction with proton pump inhibitors-related interstitial nephritis. Clin Kidney J 2023; 16:1834-1844. [PMID: 37915905 PMCID: PMC10616479 DOI: 10.1093/ckj/sfad109] [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: 01/30/2023] [Indexed: 11/03/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy and outcomes, leading to an expanding use in millions of patients worldwide. However, they can cause a spectrum of immune-related adverse events (irAEs). Essentially, any organs can be affected by irAEs, which have emerged as therapy-limiting side effects. In the kidneys, ICI-associated acute interstitial nephritis (ICI-AIN) leads to acute kidney injury (AKI) in 2%-5% of patients on ICI therapy. AKI associated with ICI therapy pathologically presents with AIN in nearly 90% of the cases, but the pathophysiology of ICI-AIN remains to be defined. The generation of autoreactive T cells in patients receiving AIN-inducible drugs, such as proton pump inhibitors (PPIs), is one of the leading theories, supported by a higher incidence of ICI-AIN in patients on these AIN-inducible drugs. In this review, we will discuss our understanding of the incidence, potential pathophysiological mechanisms, clinical presentations, risk factors, diagnosis, and management of PPI-related AIN and its interaction with ICI therapy.
Collapse
Affiliation(s)
- Jing Miao
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
42
|
Zhang SW, Wang H, Ding XH, Xiao YL, Shao ZM, You C, Gu YJ, Jiang YZ. Bidirectional crosstalk between therapeutic cancer vaccines and the tumor microenvironment: Beyond tumor antigens. FUNDAMENTAL RESEARCH 2023; 3:1005-1024. [PMID: 38933006 PMCID: PMC11197801 DOI: 10.1016/j.fmre.2022.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 11/20/2022] Open
Abstract
Immunotherapy has rejuvenated cancer therapy, especially after anti-PD-(L)1 came onto the scene. Among the many therapeutic options, therapeutic cancer vaccines are one of the most essential players. Although great progress has been made in research on tumor antigen vaccines, few phase III trials have shown clinical benefits. One of the reasons lies in obstruction from the tumor microenvironment (TME). Meanwhile, the therapeutic cancer vaccine reshapes the TME in an ambivalent way, leading to immune stimulation or immune escape. In this review, we summarize recent progress on the interaction between therapeutic cancer vaccines and the TME. With respect to vaccine resistance, innate immunosuppressive TME components and acquired resistance caused by vaccination are both involved. Understanding the underlying mechanism of this crosstalk provides insight into the treatment of cancer by directly targeting the TME or synergizing with other therapeutics.
Collapse
Affiliation(s)
- Si-Wei Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Han Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiao-Hong Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Ya-Jia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
43
|
Adeuyan O, Gordon ER, Kenchappa D, Bracero Y, Singh A, Espinoza G, Geskin LJ, Saenger YM. An update on methods for detection of prognostic and predictive biomarkers in melanoma. Front Cell Dev Biol 2023; 11:1290696. [PMID: 37900283 PMCID: PMC10611507 DOI: 10.3389/fcell.2023.1290696] [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: 09/07/2023] [Accepted: 10/04/2023] [Indexed: 10/31/2023] Open
Abstract
The approval of immunotherapy for stage II-IV melanoma has underscored the need for improved immune-based predictive and prognostic biomarkers. For resectable stage II-III patients, adjuvant immunotherapy has proven clinical benefit, yet many patients experience significant adverse events and may not require therapy. In the metastatic setting, single agent immunotherapy cures many patients but, in some cases, more intensive combination therapies against specific molecular targets are required. Therefore, the establishment of additional biomarkers to determine a patient's disease outcome (i.e., prognostic) or response to treatment (i.e., predictive) is of utmost importance. Multiple methods ranging from gene expression profiling of bulk tissue, to spatial transcriptomics of single cells and artificial intelligence-based image analysis have been utilized to better characterize the immune microenvironment in melanoma to provide novel predictive and prognostic biomarkers. In this review, we will highlight the different techniques currently under investigation for the detection of prognostic and predictive immune biomarkers in melanoma.
Collapse
Affiliation(s)
- Oluwaseyi Adeuyan
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Emily R. Gordon
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Divya Kenchappa
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Yadriel Bracero
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ajay Singh
- Albert Einstein College of Medicine, Bronx, NY, United States
| | | | - Larisa J. Geskin
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, United States
| | | |
Collapse
|
44
|
Wesevich A, Goldstein DA, Paydary K, Peer CJ, Figg WD, Ratain MJ. Interventional pharmacoeconomics for immune checkpoint inhibitors through alternative dosing strategies. Br J Cancer 2023; 129:1389-1396. [PMID: 37542109 PMCID: PMC10628132 DOI: 10.1038/s41416-023-02367-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/05/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are approved for the treatment of a variety of cancer types. The doses of these drugs, though approved by the Food and Drug Administration (FDA), have never been optimised, likely leading to significantly higher doses than required for optimal efficacy. Dose optimisation would hypothetically decrease the risk, severity, and duration of immune-related adverse events, as well as provide an opportunity to reduce costs through interventional pharmacoeconomic strategies such as off-label dose reductions or less frequent dosing. We summarise existing evidence for ICI dose optimisation to advocate for the role of interventional pharmacoeconomics.
Collapse
Affiliation(s)
- Austin Wesevich
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Daniel A Goldstein
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Clalit Health Service, Tel Aviv, Israel
- Optimal Cancer Care Alliance, Chicago, IL, USA
| | - Koosha Paydary
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Cody J Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William D Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark J Ratain
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA.
- Optimal Cancer Care Alliance, Chicago, IL, USA.
| |
Collapse
|
45
|
Boutros C, Belkadi-Sadou D, Marchand A, Roy S, Routier E, Robert C. Cured or Not? Long-term Outcomes of Immunotherapy Responders. Focus on Melanoma. Curr Oncol Rep 2023; 25:989-996. [PMID: 37266890 DOI: 10.1007/s11912-023-01429-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICIs) demonstrated robust antitumor activity and tolerable safety in advanced melanoma. Data on long-term outcome of patients who benefited from this therapy and who are still free of progression despite ICI discontinuation is now available. We review here the characteristics of long-term ICI responders and address the critical question of cure. RECENT FINDINGS Long-term outcome of patients with metastatic melanoma enrolled in large phase 2 and phase 3 clinical trials evaluating ICI in metastatic melanoma is now available. Durable responses, with more than 6 years of median follow-up, may persist after discontinuation. They occur more frequently in patients who achieved a complete response rather than in patients who had partial response or stable disease. Although long-term clinical benefit is more frequent in patients with high PDL-1 expression and smaller tumor burden, durable response may also be observed regardless of baseline characteristics. In patients with asymptomatic brain metastasis, combined immunotherapy (ipilimumab plus nivolumab) may also lead to long-term remission. Clinical trials confirm the durable antitumor activity of ICI. Although the hope for cure seems reasonable for many patients in this situation, late relapses may occur and no relapse-predictive biomarkers have been identified yet. Long-term responders who relapse can respond to a rechallenge of ICI although data are limited concerning the rate and the duration of this new response.
Collapse
Affiliation(s)
- Céline Boutros
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
- Outpatient Clinic, Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Djaouida Belkadi-Sadou
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Antoine Marchand
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Séverine Roy
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Emilie Routier
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Caroline Robert
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
- University Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France.
- INSERM Unit U981, Villejuif, France.
| |
Collapse
|
46
|
Kovács SA, Fekete JT, Győrffy B. Predictive biomarkers of immunotherapy response with pharmacological applications in solid tumors. Acta Pharmacol Sin 2023; 44:1879-1889. [PMID: 37055532 PMCID: PMC10462766 DOI: 10.1038/s41401-023-01079-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Immune-checkpoint inhibitors show promising effects in the treatment of multiple tumor types. Biomarkers are biological indicators used to select patients for a systemic anticancer treatment, but there are only a few clinically useful biomarkers such as PD-L1 expression and tumor mutational burden, which can be used to predict immunotherapy response. In this study, we established a database consisting of both gene expression and clinical data to identify biomarkers of response to anti-PD-1, anti-PD-L1, and anti-CTLA-4 immunotherapies. A GEO screening was executed to identify datasets with simultaneously available clinical response and transcriptomic data regardless of cancer type. The screening was restricted to the studies involving administration of anti-PD-1 (nivolumab, pembrolizumab), anti-PD-L1 (atezolizumab, durvalumab) or anti-CTLA-4 (ipilimumab) agents. Receiver operating characteristic (ROC) analysis and Mann-Whitney test were executed across all genes to identify features related to therapy response. The database consisted of 1434 tumor tissue samples from 19 datasets with esophageal, gastric, head and neck, lung, and urothelial cancers, plus melanoma. The strongest druggable gene candidates linked to anti-PD-1 resistance were SPIN1 (AUC = 0.682, P = 9.1E-12), SRC (AUC = 0.667, P = 5.9E-10), SETD7 (AUC = 0.663, P = 1.0E-09), FGFR3 (AUC = 0.657, P = 3.7E-09), YAP1 (AUC = 0.655, P = 6.0E-09), TEAD3 (AUC = 0.649, P = 4.1E-08) and BCL2 (AUC = 0.634, P = 9.7E-08). In the anti-CTLA-4 treatment cohort, BLCAP (AUC = 0.735, P = 2.1E-06) was the most promising gene candidate. No therapeutically relevant target was found to be predictive in the anti-PD-L1 cohort. In the anti-PD-1 group, we were able to confirm the significant correlation with survival for the mismatch-repair genes MLH1 and MSH6. A web platform for further analysis and validation of new biomarker candidates was set up and available at https://www.rocplot.com/immune . In summary, a database and a web platform were established to investigate biomarkers of immunotherapy response in a large cohort of solid tumor samples. Our results could help to identify new patient cohorts eligible for immunotherapy.
Collapse
Affiliation(s)
- Szonja Anna Kovács
- Department of Bioinformatics, Semmelweis University, Tűzoltó utca 7-9, 1094, Budapest, Hungary
- Doctoral School of Pathological Sciences, Semmelweis University, Üllői út 26, 1085, Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar tudósok körútja 2 1117, Budapest, Hungary
| | - János Tibor Fekete
- National Laboratory for Drug Research and Development, Magyar tudósok körútja 2 1117, Budapest, Hungary
- Research Centre for Natural Sciences, Oncology Biomarker Research Group, Institute of Enzymology, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, 1117, Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Tűzoltó utca 7-9, 1094, Budapest, Hungary.
- Department of Pediatrics, Semmelweis University, Tűzoltó utca 7-9, 1094, Budapest, Hungary.
| |
Collapse
|
47
|
Rui R, Zhou L, He S. Cancer immunotherapies: advances and bottlenecks. Front Immunol 2023; 14:1212476. [PMID: 37691932 PMCID: PMC10484345 DOI: 10.3389/fimmu.2023.1212476] [Citation(s) in RCA: 133] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/26/2023] [Indexed: 09/12/2023] Open
Abstract
Immunotherapy has ushered in a new era in cancer treatment, and cancer immunotherapy continues to be rejuvenated. The clinical goal of cancer immunotherapy is to prime host immune system to provide passive or active immunity against malignant tumors. Tumor infiltrating leukocytes (TILs) play an immunomodulatory role in tumor microenvironment (TME) which is closely related to immune escape of tumor cells, thus influence tumor progress. Several cancer immunotherapies, include immune checkpoint inhibitors (ICIs), cancer vaccine, adoptive cell transfer (ACT), have shown great efficacy and promise. In this review, we will summarize the recent research advances in tumor immunotherapy, including the molecular mechanisms and clinical effects as well as limitations of immunotherapy.
Collapse
Affiliation(s)
- Rui Rui
- Department of Urology, Peking University First Hospital, Beijing, China
- The Institution of Urology, Peking University, Beijing, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, China
- National Urological Cancer Center, Beijing, China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing, China
- The Institution of Urology, Peking University, Beijing, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, China
- National Urological Cancer Center, Beijing, China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Beijing, China
- The Institution of Urology, Peking University, Beijing, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, China
- National Urological Cancer Center, Beijing, China
| |
Collapse
|
48
|
Ocaña-Guzmán R, Osorio-Pérez D, Chavez-Galan L. Opportunistic Infections and Immune-Related Adverse Events Associated with Administering Immune Checkpoint Inhibitors: A Narrative Review. Pharmaceuticals (Basel) 2023; 16:1119. [PMID: 37631034 PMCID: PMC10458516 DOI: 10.3390/ph16081119] [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: 06/27/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Manipulating the immune system by blocking the immune checkpoint receptors is the basis of immunotherapy, a relevant tool in current clinical oncology. The strategy of blocking the immune checkpoints (Immune Checkpoint Inhibitors, ICI) consists of using monoclonal antibodies to inhibit the interaction between ligand and inhibitory receptors from triggering a complete activation of helper and cytotoxic T cells to fight against tumour cells. Immunotherapy has benefited patients with diverse cancers such as stomach, lung, melanoma, and head and neck squamous cell carcinoma, among others. Unfortunately, a growing number of reports have indicated that the ICI treatment also can show a dark side under specific conditions; some of the adverse effects induced by ICI are immunosuppression, opportunistic infections, and organ-specific alterations. This review discusses some immunologic aspects related to these unwanted effects.
Collapse
Affiliation(s)
- Ranferi Ocaña-Guzmán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico;
| | - Diego Osorio-Pérez
- Department of Medical Oncology, Hospital de la Mujer, Mexico City 11340, Mexico;
| | - Leslie Chavez-Galan
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico;
| |
Collapse
|
49
|
Danielsen JT, Strøm L, Knutzen SM, Schmidt H, Amidi A, Wu LM, Zachariae R. Psychological and behavioral symptoms in patients with melanoma: A systematic review and meta-analysis. Psychooncology 2023; 32:1208-1222. [PMID: 37370196 DOI: 10.1002/pon.6184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/12/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE Improved survival rates have made it increasingly important for clinicians to focus on cancer survivorship issues affecting the quality of life of melanoma patients. To provide a comprehensive overview of the disease and treatment-related issues affecting such patients, we conducted a systematic review and meta-analysis of the literature to estimate the prevalence of symptoms of depression, anxiety, fatigue, sleep disturbance, and cognitive problems among melanoma patients, both uveal and cutaneous, before, during and after treatment. METHODS The review was preregistered with PROSPERO (#CRD42020189847) and reported following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. A comprehensive search of the literature published up until June 2022 was undertaken using PubMed, PsycInfo, the Cochrane Library, and CINAHL. Two independent reviewers screened 1418 records and quality-rated included studies. The reported prevalence rates of symptoms were pooled using a random-effects model. RESULTS Sixty-six studies including a total of 12,400 melanoma patients published between 1992 and 2022 were included. Pooled prevalence rates ranged from 6% to 16% for depression and 7%-30% for anxiety across diagnoses (uveal and cutaneous melanoma) and assessment time points. One third of the patients (35%) reported clinically significant fatigue, 20%-44% had cognitive complaints, while prevalence of sleep disturbance was not reported. Quality assessment indicated that 80% of the studies were of good quality. CONCLUSION A large body of research shows that depression and anxiety symptoms are prevalent in melanoma patients before, during and after treatment. However, research examining other symptoms known to affect quality of life, such as fatigue, sleep disturbances, and cognitive problems, is still needed.
Collapse
Affiliation(s)
- Josefine T Danielsen
- Unit for Psycho-Oncology & Health Psychology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Psychology and Behavioural Sciences, Aarhus University, Aarhus, Denmark
| | - Louise Strøm
- Unit for Psycho-Oncology & Health Psychology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Psychology and Behavioural Sciences, Aarhus University, Aarhus, Denmark
| | - Sofie M Knutzen
- Unit for Psycho-Oncology & Health Psychology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Psychology and Behavioural Sciences, Aarhus University, Aarhus, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Ali Amidi
- Unit for Psycho-Oncology & Health Psychology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Psychology and Behavioural Sciences, Aarhus University, Aarhus, Denmark
| | - Lisa M Wu
- Unit for Psycho-Oncology & Health Psychology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Psychology and Behavioural Sciences, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Robert Zachariae
- Unit for Psycho-Oncology & Health Psychology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Psychology and Behavioural Sciences, Aarhus University, Aarhus, Denmark
| |
Collapse
|
50
|
Ji Z, Shen J, Lan Y, Yi Q, Liu H. Targeting signaling pathways in osteosarcoma: Mechanisms and clinical studies. MedComm (Beijing) 2023; 4:e308. [PMID: 37441462 PMCID: PMC10333890 DOI: 10.1002/mco2.308] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 07/15/2023] Open
Abstract
Osteosarcoma (OS) is a highly prevalent bone malignancy among adolescents, accounting for 40% of all primary malignant bone tumors. Neoadjuvant chemotherapy combined with limb-preserving surgery has effectively reduced patient disability and mortality, but pulmonary metastases and OS cells' resistance to chemotherapeutic agents are pressing challenges in the clinical management of OS. There has been an urgent need to identify new biomarkers for OS to develop specific targeted therapies. Recently, the continued advancements in genomic analysis have contributed to the identification of clinically significant molecular biomarkers for diagnosing OS, acting as therapeutic targets, and predicting prognosis. Additionally, the contemporary molecular classifications have revealed that the signaling pathways, including Wnt/β-catenin, PI3K/AKT/mTOR, JAK/STAT3, Hippo, Notch, PD-1/PD-L1, MAPK, and NF-κB, have an integral role in OS onset, progression, metastasis, and treatment response. These molecular classifications and biological markers have created new avenues for more accurate OS diagnosis and relevant treatment. We herein present a review of the recent findings for the modulatory role of signaling pathways as possible biological markers and treatment targets for OS. This review also discusses current OS therapeutic approaches, including signaling pathway-based therapies developed over the past decade. Additionally, the review covers the signaling targets involved in the curative effects of traditional Chinese medicines in the context of expression regulation of relevant genes and proteins through the signaling pathways to inhibit OS cell growth. These findings are expected to provide directions for integrating genomic, molecular, and clinical profiles to enhance OS diagnosis and treatment.
Collapse
Affiliation(s)
- Ziyu Ji
- School of Integrated Traditional Chinese and Western MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Jianlin Shen
- Department of OrthopaedicsAffiliated Hospital of Putian UniversityPutianFujianChina
| | - Yujian Lan
- School of Integrated Traditional Chinese and Western MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Qian Yi
- Department of PhysiologySchool of Basic Medical ScienceSouthwest Medical UniversityLuzhouSichuanChina
| | - Huan Liu
- Department of OrthopaedicsThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouSichuanChina
| |
Collapse
|