601
|
Lethal form of a late-onset aquaporin-4 antibody-positive NMOSD related to the immune checkpoint inhibitor nivolumab. J Neurol 2021; 269:2778-2780. [PMID: 34841443 PMCID: PMC9021101 DOI: 10.1007/s00415-021-10913-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022]
|
602
|
Bai Y, Sha J, Okui T, Moriyama I, Ngo HX, Tatsumi H, Kanno T. The Epithelial-Mesenchymal Transition Influences the Resistance of Oral Squamous Cell Carcinoma to Monoclonal Antibodies via Its Effect on Energy Homeostasis and the Tumor Microenvironment. Cancers (Basel) 2021; 13:5905. [PMID: 34885013 PMCID: PMC8657021 DOI: 10.3390/cancers13235905] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/12/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a major type of cancer that accounts for over 90% of all oral cancer cases. Recently developed evidence-based therapeutic regimens for OSCC based on monoclonal antibodies (mAbs), such as cetuximab, pembrolizumab, and nivolumab, have attracted considerable attention worldwide due to their high specificity, low toxicity, and low rates of intolerance. However, the efficacy of those three mAbs remains poor because of the low rate of responders and acquired resistance within a short period of time. The epithelial-mesenchymal transition (EMT) process is fundamental for OSCC growth and metastasis and is also responsible for the poor response to mAbs. During EMT, cancer cells consume abundant energy substrates and create an immunosuppressive tumor microenvironment to support their growth and evade T cells. In this review, we provide an overview of the complex roles of major substrates and signaling pathways involved in the development of therapeutic resistance in OSCC. In addition, we summarize potential therapeutic strategies that may help overcome this resistance. This review aims to help oral oncologists and researchers aiming to manage OSCC and establish new treatment modalities.
Collapse
Affiliation(s)
- Yunpeng Bai
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Jingjing Sha
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Tatsuo Okui
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Ichiro Moriyama
- Department of Medical Oncology/Innovative Cancer Center, Shimane University Hospital, Izumo, Shimane 693-8501, Japan;
| | - Huy Xuan Ngo
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Hiroto Tatsumi
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Takahiro Kanno
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| |
Collapse
|
603
|
Treatment-Related Adverse Events with PD-1 or PD-L1 Inhibitors: A Systematic Review and Meta-Analysis. Life (Basel) 2021; 11:life11111277. [PMID: 34833153 PMCID: PMC8618590 DOI: 10.3390/life11111277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
Objective: to evaluate the risk of treatment-related adverse events of different severity and different system with PD-1 or PD-L1 inhibitors. Methods: randomized controlled trials (RCTs) that using PD-1/PD-L1 for cancer treatment were searched in the PubMed, Embase, Cochrane Library, and Web of Science from 1 January 2019 to 31 May 2021. Adverse events data were extracted from clinical trials website or original article by two authors separately. Meta-analysis was used to determine risk ratio (RR) and 95% confidence interval (95% CI) of adverse events in PD-1/PD-L1 inhibitors groups compared to that of control groups. Subgroup analyses were also performed. Results: a total of 5,807 studies were initially identified and after exclusion, 41 studies were included in meta-analysis. All the trials were international multicenter, randomized, phase II/III clinical trials, with the median follow-up of 27.5 months on average. Analysis of all grade adverse events showed that PD-1/PD-L1 inhibitors treatment significantly increased the risk of immune-related adverse events, including pruritus (RR: 2.34, 95% CI: 1.85–2.96), rash (RR: 1.53, 95% CI: 1.25–1.87), ALT elevation (RR 1.54, 95% CI 1.23–1.92), AST elevation (AST: RR 1.49, 95% CI 1.20–1.85), hepatitis (RR: 3.54, 95% CI: 1.96–6.38) and hypothyroid (RR: 5.29, 95% CI: 4.00–6.99) compared with that of control group. Besides that, PD-1/PD-L1 inhibitors were associated with higher risk of adverse events related to respiratory system including cough (RR: 1.33, 95% CI: 1.21–1.48), dyspnea (RR:1.23, 95% CI: 1.12–1.35) and chest pain (RR: 1.26, 95% CI: 1.07–1.47) compared with that of control groups in our meta-analysis and the dyspnea was taken high risk both in all grade and grade 3 or higher (RR: 1.55, 95% CI: 1.13–2.12). The risk of arthralgia was increased with PD-1/PD-L1 inhibitors (RR: 1.27, 95% CI: 1.10–1.47). Although the risk of myalgia was similar with PD-1/PD-L1 inhibitors and control groups, under subgroup analysis, PD-1/PD-L1 inhibitors decreased the risk of myalgia (RR: 0.56, 95% CI: 0.45–0.70) compared with that of chemotherapy. Conclusions: our results provide clear evidence that the risk of treatment-related adverse events in PD-1 or PD-L1 varies widely in different system. In particular, when using PD-1/PD-L1 inhibitors for oncology treatment, besides the common immune-related adverse events like pruritus, rash, hepatitis, and hypothyroid, the respiratory disorders and musculoskeletal disorders, such as cough, dyspnea, arthralgia, and myalgia, should also be taken into consideration.
Collapse
|
604
|
Petruk A, Kamyshna I, Shkilna M, Kamyshnyi A. Glucocorticoid-induced Changes in the Transcriptional Activity of Genes of the Innate and Adaptive Immune System in the Blood of Patients with Acute Urticaria. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: A number of the main effects of glucocorticoids (GCs) are their direct action on T cells, mainly through the transcriptional regulation: elevated expression of immune-regulatory proteins, inhibitory receptors, and reduced expression of pro-inflammatory cytokines, co-stimulatory molecules, and cell cycle mediators. But controversies arise due to the clinical effectiveness of GCs in the treatment of acute urticaria.
Methods: In our research, we applied a pathway-specific PCR array (Human Innate & Adaptive Immune Responses RT2 Profiler PCR Array, QIAGEN, Germany) to detect and verify innate & adaptive immune responses pathway-focused genes expression in the blood of patients with acute urticaria who received treatment with glucocorticoids in addition to standard therapy.
Results: Adding glucocorticoids to standard therapy did not notably affect the nature of the clinical presentation of acute urticaria, which was assessed according to the UAS scale (urticaria activity score). Analysis of the transcriptional profile of peripheral blood mononuclear cells in patients with acute urticaria against the background of glucocorticoid therapy showed the induction expression of the FOXP3 and IL10 genes against the background of repression of the transcriptional activity of the genes for chemokines and cytokines CCL5, CXCL8, IFNG, IL2, IL5, IL17A, IL1B, and TNF. Glucocorticoid-induced changes in the transcriptome also manifested by pronounced repression in genes of CD40 and CD80 (B7-1) co-stimulatory molecules, transcriptional regulators of Th1-cells differentiation - TBX21 and STAT1, Th17 cells - RORC, NLRP3-inflammasome genes, and the transcription factor NFKB1 compared with the control group.
Conclusions: Adding glucocorticoids to the standard therapy of acute urticaria has a pronounced immunosuppressive potential at the transcriptome level of immune response genes in the blood; however, it does not have any noticeable clinical effect.
Collapse
|
605
|
Zavadinack M, de Lima Bellan D, da Rocha Bertage JL, da Silva Milhorini S, da Silva Trindade E, Simas FF, Sassaki GL, Cordeiro LMC, Iacomini M. An α-D-galactan and a β-D-glucan from the mushroom Amanita muscaria: Structural characterization and antitumor activity against melanoma. Carbohydr Polym 2021; 274:118647. [PMID: 34702466 DOI: 10.1016/j.carbpol.2021.118647] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/03/2021] [Accepted: 09/04/2021] [Indexed: 12/13/2022]
Abstract
Polysaccharides α-D-galactan (GAL-Am) and β-D-glucan (GLC-Am) were obtained from Amanita muscaria fruiting bodies. They were purified using different methodologies, such as Fehling precipitation (for both fractions), freeze-thawing process and ultrafiltration (for GLC-Am). Results showed that the GAL-Am has (1 → 6)-linked Galp main chain branched at O-2 by terminal Galp units and has not been previously reported. Besides, GLC-Am has (1 → 3)-linked Glcp in the main chain, substituted at O-6 by (1 → 6)-linked β-Glcp units. Both are water-soluble, with 9.0 × 103 g/moL and 1.3 × 105 g/moL, respectively. GAL-Am and GLC-Am presented a selective proliferation reduction against B16-F10 melanoma cell line, not affecting non tumoral BALB/3T3 fibroblast cell line. Furthermore, both fractions reduced clonogenic capacity of melanoma cell line over an extended period of time. These results were obtained without modulations in B16-F10 cell adhesion, reinforcing the biological activities towards cell proliferation impairment and eliciting these polysaccharides as promising compounds to further exploration of their antimelanoma properties.
Collapse
Affiliation(s)
- Matheus Zavadinack
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR CEP 81531-980, Brazil
| | - Daniel de Lima Bellan
- Department of Cell Biology, Federal University of Paraná, Curitiba, PR CEP 81531-980, Brazil
| | | | - Shayane da Silva Milhorini
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR CEP 81531-980, Brazil
| | | | | | - Guilherme Lanzi Sassaki
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR CEP 81531-980, Brazil
| | - Lucimara M C Cordeiro
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR CEP 81531-980, Brazil
| | - Marcello Iacomini
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR CEP 81531-980, Brazil.
| |
Collapse
|
606
|
Personeni N, Pressiani T, D’Alessio A, Prete MG, Bozzarelli S, Terracciano L, Dal Buono A, Capogreco A, Aghemo A, Lleo A, Lutman RF, Roncalli M, Giordano L, Santoro A, Di Tommaso L, Rimassa L. Hepatotoxicity in Patients with Hepatocellular Carcinoma on Treatment with Immune Checkpoint Inhibitors. Cancers (Basel) 2021; 13:cancers13225665. [PMID: 34830823 PMCID: PMC8616285 DOI: 10.3390/cancers13225665] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Hepatitis is a relatively frequent immune-related adverse event in patients with hepatocellular carcinoma receiving immunotherapy, but risk factors and clinical course are unclear. Herein, we show that the development of high-grade hepatitis is associated with increased baseline ALT levels and infectious etiology of hepatocellular carcinoma (related to prior hepatitis B or C virus exposure). In addition, when resolved, high-grade hepatitis does not preclude treatment resumption and does not affect subsequent time to treatment failure. Analysis of baseline tumor specimens, at a preliminary level, suggests that biological features reminiscent of the hepatocellular carcinoma “immune class” could protect against high-grade hepatitis development, thereby warranting further investigation. Abstract Risk factors for hepatic immune-related adverse events (HIRAEs) in patients with advanced/unresectable hepatocellular carcinoma (HCC) treated with immune checkpoint inhibitors (ICIs) are unclear. We investigated: (i) clinical and morpho-pathological predictors of HIRAEs in 27 pretreatment tumor specimens, including surrogate biomarkers of the HCC immune class (based on intratumoral tertiary lymphoid structures, and glutamine synthase, CD3, and CD79 expression); and (ii) the relationship between HIRAE onset and subsequent treatment outcomes. Fifty-eight patients were included—20 (34%) received ICIs alone, and 38 (66%) received ICIs plus targeted agents as first- or further-line treatment. After a median time of 0.9 months (range, 0.4–2.7), nine patients (15.5%) developed grade ≥ 3 hepatitis, which was significantly associated with higher baseline ALT levels (p = 0.037), and an infectious HCC etiology (p = 0.023). ICIs were safely resumed in six out of nine patients. Time to treatment failure (TTF) was not significantly different in patients developing grade ≥ 3 hepatitis vs. lower grades (3.25 vs. 3.91 months, respectively; p = 0.81). Biomarker surrogates for the HCC immune class were not detected in patients developing grade ≥ 3 hepatitis. Grade ≥ 3 hepatitis has a benign course that does not preclude safe ICI reintroduction, without any detrimental effect on TTF.
Collapse
Affiliation(s)
- Nicola Personeni
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (T.P.); (S.B.); (L.G.)
| | - Tiziana Pressiani
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (T.P.); (S.B.); (L.G.)
| | - Antonio D’Alessio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
| | - Maria Giuseppina Prete
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
| | - Silvia Bozzarelli
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (T.P.); (S.B.); (L.G.)
| | - Luigi Terracciano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Pathology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Arianna Dal Buono
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Antonio Capogreco
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Alessio Aghemo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Romano Fabio Lutman
- Department of Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy;
| | - Massimo Roncalli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Pathology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Laura Giordano
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (T.P.); (S.B.); (L.G.)
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (T.P.); (S.B.); (L.G.)
| | - Luca Di Tommaso
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Pathology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (N.P.); (A.D.); (M.G.P.); (L.T.); (A.D.B.); (A.C.); (A.A.); (A.L.); (M.R.); (A.S.); (L.D.T.)
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (T.P.); (S.B.); (L.G.)
- Correspondence: ; Tel.: +39-02-82244573; Fax: +39-02-82244590
| |
Collapse
|
607
|
Harnessing the combined potential of cancer immunotherapy and nanomedicine: A new paradigm in cancer treatment. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 40:102492. [PMID: 34775062 DOI: 10.1016/j.nano.2021.102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/16/2021] [Accepted: 10/29/2021] [Indexed: 11/21/2022]
Abstract
Cancer immunotherapy has recently emerged as a rising star due to its ability to activate patients' immune systems to fight tumors and prevent relapse. Conversely, the interest in cancer nanomedicine has seemingly waned due to its lackluster clinical translation. Despite being hailed as a game-changer in oncology, cancer immunotherapy still faces numerous challenges. Combining both entities together has thus been one among several solutions proposed to circumvent these challenges. This solution has since gained traction and has also led to a renaissance of cancer nanomedicine. While most combinations are currently experimental at best, some have progressed on to clinical trials. This review thus seeks to examine the advantages and disadvantages of integrating both modalities as a cancer treatment. The opportunities, challenges and future directions of this emerging field will also be explored with the hope that such a combination will lead to a paradigm shift in cancer treatments.
Collapse
|
608
|
Sun YM, Li W, Chen ZY, Wang Y. Risk of Pneumonitis Associated With Immune Checkpoint Inhibitors in Melanoma: A Systematic Review and Network Meta-Analysis. Front Oncol 2021; 11:651553. [PMID: 34745932 PMCID: PMC8568299 DOI: 10.3389/fonc.2021.651553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have dramatically altered the treatment landscape for patients with melanoma. However, their use also generates unique immune-related adverse effects (irAEs). We performed a systematic review and network meta‐analysis to compare the risk of pneumonitis associated with ICIs for patients with advanced or metastatic melanoma. Methods Phase II/III randomized clinical trials (RCTs) with ICIs were identified through comprehensive searches of multiple databases. An NMA was conducted to compare the risk of pneumonitis associated with ICIs and all‐grade (grade 1‐5) and high‐grade (grade 3‐5) immune‐related pneumonitis (IRP) were estimated by odds ratios (ORs). Results A total of 10 randomized clinical trials involving 5,335 patients were enrolled in this study. Conventional chemotherapy was associated with a lower risk of grade 1–5 IRP compared with ICIs monotherapy (OR, 0.14, 95% CI, 0.03 to 0.73) and dual ICIs combination (OR, 0.03, 95% CI, 0.00 to 0.19). In addition, dual ICIs combination showed a noticeably higher risk than ICI monotherapy (OR, 4.45, 95% CI, 2.14 to 9.25) of grade 1–5 IRP. No significant difference in grade 1–5 IRP was observed between cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) inhibitors. As to grade 3‐5 IRP, no statistically significant difference was found among different ICIs-based regimens. Conclusion These findings revealed that ICIs could increase the risk of all-grade pneumonitis for patients with advanced melanoma, compared with conventional chemotherapy. Dual ICIs combination could further increase the risk of all-grade pneumonitis than ICIs monotherapy. There was no significant difference in the risk of pneumonia between CTLA-4 and PD-1 inhibitors.
Collapse
Affiliation(s)
- You-Meng Sun
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Li
- School of Statistics and Mathematics, Zhejiang Gongshang University, Hangzhou, China
| | - Zhi-Yu Chen
- School of Statistics and Mathematics, Zhejiang Gongshang University, Hangzhou, China
| | - Ying Wang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
609
|
Pérez Santamaría C, Castillo-López G, Curto-García I, Gómez Iglesias P, Lumbreras Cabrera M, Carreira Guillán L. Severe pembrolizumab-induced gastritis in a patient with lung adenocarcinoma. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2021; 114:127-128. [PMID: 34743523 DOI: 10.17235/reed.2021.8319/2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cancer immunotherapy is an increasingly common treatment option based on anti-tumor immune response. However, a challenge is arising regarding potential, as yet poorly recognized immune-mediated adverse effects.
Collapse
|
610
|
The Price of Success: Immune-Related Adverse Events from Immunotherapy in Lung Cancer. Curr Oncol 2021; 28:4392-4407. [PMID: 34898551 PMCID: PMC8628657 DOI: 10.3390/curroncol28060373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/19/2021] [Accepted: 10/31/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy has the goal of enhancing a patient’s intrinsic immune processes in order to mount a successful immune response against tumor cells. Cancer cells actively employ tactics to evade, delay, alter, or attenuate the anti-tumor immune response. Immune checkpoint inhibitors (ICIs) modulate endogenous regulatory immune mechanisms to enhance immune system activation, and have become the mainstay of therapy in many cancer types. This activation occurs broadly and as a result, activation is supraphysiologic and relatively non-specific, which can lead to immune-related adverse events (irAEs), the frequency of which depends on the patient, the cancer type, and the specific ICI antibody. Careful assessment of patients for irAEs through history taking, physical exam, and routine laboratory assessments are key to identifying irAEs at early stages, when they can potentially be managed more easily and before progressing to higher grades or more serious effects. Generally, most patients with low grade irAEs are eligible for re-challenge with ICIs, and the use of corticosteroids to address an irAE is not associated with poorer patient outcomes. This paper reviews immune checkpoint inhibitors (ICIs) including their mechanisms of action, usage, associated irAEs, and their management.
Collapse
|
611
|
Zhou J, Mao Q, Li Y, Li Z, He H, Chen Q, Liu C. Oral reactive capillary hemangiomas induced by SHR-1210 in the treatment of non-small cell lung cancer: a case report and literature review. BMC Oral Health 2021; 21:559. [PMID: 34727912 PMCID: PMC8561900 DOI: 10.1186/s12903-021-01901-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antibodies to PD-1 and PD-L1 have remarkably improved the overall survival of many patients with advanced solid tumors. SHR-1210 is an anti-PD-1 monoclonal antibody. Dermatologic reactive capillary hemangiomas (RCH) were the most common and unique drug-related AEs of SHR-1210, but rare on oral mucosa and gastrointestinal mucosa. Herein we report a case of RCH occurred in oral mucosa during the clinical trials of SHR-1210 in the treatment of non-small cell lung cancer. CASE PRESENTATION A male in his 60 s with a history of non-small cell lung cancer received injection of anti-PD-1 monoclonal antibodies SHR-1210. The patient developed drug-related RCH on skin after the first injection and began to have gingival hyperplasia one year after the first injection which gradually increased in size and affect eating and speaking. Anti-PD-1 treatments were continued. After periodontal treatment, two oral lesions and one skin lesion were surgically removed. Similar histological manifestation was found in all three lesions as reactive capillary hemangiomas. All lesions had a good prognosis without recurrence on oral mucosa within one year after surgery. CONCLUSIONS Oral reactive capillary hemangiomas could be induced by SHR-1210 in the treatment of non-small cell lung cancer. Surgical resection is an effective treatment with a good prognosis.
Collapse
Affiliation(s)
- Jinhan Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medcine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Rsearch of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Qinghua Mao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medcine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Rsearch of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Yining Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medcine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Rsearch of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Zhiyong Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medcine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Rsearch of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Hong He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medcine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Rsearch of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medcine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Rsearch of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Chuanxia Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medcine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Rsearch of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
612
|
Yang T, Mochida Y, Liu X, Zhou H, Xie J, Anraku Y, Kinoh H, Cabral H, Kataoka K. Conjugation of glucosylated polymer chains to checkpoint blockade antibodies augments their efficacy and specificity for glioblastoma. Nat Biomed Eng 2021; 5:1274-1287. [PMID: 34635819 DOI: 10.1038/s41551-021-00803-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 09/01/2021] [Indexed: 02/07/2023]
Abstract
Because of the blood-tumour barrier and cross-reactivity with healthy tissues, immune checkpoint blockade therapy against glioblastoma has inadequate efficacy and is associated with a high risk of immune-related adverse events. Here we show that anti-programmed death-ligand 1 antibodies conjugated with multiple poly(ethylene glycol) (PEG) chains functionalized to target glucose transporter 1 (which is overexpressed in brain capillaries) and detaching in the reductive tumour microenvironment augment the potency and safety of checkpoint blockade therapy against glioblastoma. In mice bearing orthotopic glioblastoma tumours, a single dose of glucosylated and multi-PEGylated antibodies reinvigorated antitumour immune responses, induced immunological memory that protected the animals against rechallenge with tumour cells, and suppressed autoimmune responses in the animals' healthy tissues. Drug-delivery formulations leveraging multivalent ligand interactions and the properties of the tumour microenvironment to facilitate the crossing of blood-tumour barriers and increase drug specificity may enhance the efficacy and safety of other antibody-based therapies.
Collapse
Affiliation(s)
- Tao Yang
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Yuki Mochida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Xueying Liu
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Hang Zhou
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Jinbing Xie
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Yasutaka Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Hiroaki Kinoh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan. .,Institute for Future Initiatives, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
613
|
Xie Y, Xie F, Zhang L, Zhou X, Huang J, Wang F, Jin J, Zhang L, Zeng L, Zhou F. Targeted Anti-Tumor Immunotherapy Using Tumor Infiltrating Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101672. [PMID: 34658167 PMCID: PMC8596143 DOI: 10.1002/advs.202101672] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/21/2021] [Indexed: 05/08/2023]
Abstract
In the tumor microenvironment, T cells, B cells, and many other cells play important and distinct roles in anti-tumor immunotherapy. Although the immune checkpoint blockade and adoptive cell transfer can elicit durable clinical responses, only a few patients benefit from these therapies. Increased understanding of tumor-infiltrating immune cells can provide novel therapies and drugs that induce a highly specific anti-tumor immune response to certain groups of patients. Herein, the recent research progress on tumor-infiltrating B cells and T cells, including CD8+ T cells, CD4+ T cells, and exhausted T cells and their role in anti-tumor immunity, is summarized. Moreover, several anti-tumor therapy approaches are discussed based on different immune cells and their prospects for future applications in cancer treatment.
Collapse
Affiliation(s)
- Yifan Xie
- School of MedicineZhejiang University City CollegeHangzhou310015China
- College of Life SciencesZhejiang UniversityHangzhou310058China
| | - Feng Xie
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Lei Zhang
- Department of Orthopaedic SurgeryThe Third Affiliated Hospital of Wenzhou Medical UniversityRui'an325200China
| | - Xiaoxue Zhou
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Jun Huang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Fangwei Wang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Jin Jin
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Long Zhang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Linghui Zeng
- School of MedicineZhejiang University City CollegeHangzhou310015China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| |
Collapse
|
614
|
Krusche M, Schneider U, Geisler C, Keller S, Stenzel W, Ohrndorf S. [Myofasciitis under nivolumab treatment]. Z Rheumatol 2021; 80:884-888. [PMID: 33885949 PMCID: PMC8575749 DOI: 10.1007/s00393-021-01001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 11/20/2022]
Abstract
We report the case of a 73-year-old female patient with malignant melanoma who developed rapidly progressive dermatosclerosis of the arms and legs as well as myalgia and flexion contractures during treatment with the immune checkpoint inhibitor nivolumab. The diagnosis of a myofasciitis was confirmed by imaging and biopsy. Following consultation with the treating dermato-oncologists nivolumab treatment was paused and treatment with methotrexate and prednisolone was initiated. Immune checkpoint inhibitors can induce a variety of immune-mediated side effects and can also imitate symptoms of rheumatological diseases. The occurrence of myofasciitis under immune checkpoint inhibition has been reported in the literature only in a few cases. Further oncological and rheumatological treatment management should be carried out in close interdisciplinary coordination.
Collapse
Affiliation(s)
- M Krusche
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin, Charitéplatz 1, 10117, Berlin, Deutschland.
| | - U Schneider
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin, Charitéplatz 1, 10117, Berlin, Deutschland
| | - C Geisler
- Klinik für Dermatologie, Venerologie und Allergologie, Charité - Universitätsmedizin, Charitéplatz 1, 10117, Berlin, Deutschland
| | - S Keller
- Klinik für Radiologie, Charité - Universitätsmedizin, Charitéplatz 1, 10117, Berlin, Deutschland
| | - W Stenzel
- Institut für Neuropathologie, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland
| | - S Ohrndorf
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin, Charitéplatz 1, 10117, Berlin, Deutschland
| |
Collapse
|
615
|
Asan MF, Castelino RL, Babu SG, Rao K, Pandita V. Oral Immune-Related Adverse Events - Current Concepts and their Management. Asia Pac J Oncol Nurs 2021; 8:604-609. [PMID: 34790844 PMCID: PMC8522587 DOI: 10.4103/apjon.apjon-2136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/10/2021] [Indexed: 11/05/2022] Open
Abstract
Recent advent of immune checkpoint inhibitors (ICIs) have made significant improvement in the treatment outcome of cancer patients. They are also known to increase the overall survival in many malignancies. They target key immune checkpoints, acting on the cytotoxic T-lymphocyte antigen-4, programmed death-1 (PD-1), and PD-1 ligand 1 pathways. ICIs are effective in cancer therapy, but also possess various adverse effects that are termed together as immune-related adverse events (irAEs). Information focusing only on the oral reactions of irAEs is scanty in the literature. Therefore, we performed a computerized database search in PubMed and Google Scholar to identify and collect data regarding the oral adverse effects of ICIs. The early recognition of oral irAEs and appropriative intervention may help in improving the quality of life in patients. This paper presents a brief review of oral irAEs and their management.
Collapse
Affiliation(s)
- Mohamed Faizal Asan
- Department of Oral Medicine and Radiology, Nitte (Deemed to be University), AB Shetty Memorial Institute of Dental Sciences, Mangalore, Karnataka, India
| | - Renita Lorina Castelino
- Department of Oral Medicine and Radiology, Nitte (Deemed to be University), AB Shetty Memorial Institute of Dental Sciences, Mangalore, Karnataka, India
| | - Subhas G. Babu
- Department of Oral Medicine and Radiology, Nitte (Deemed to be University), AB Shetty Memorial Institute of Dental Sciences, Mangalore, Karnataka, India
| | - Kumuda Rao
- Department of Oral Medicine and Radiology, Nitte (Deemed to be University), AB Shetty Memorial Institute of Dental Sciences, Mangalore, Karnataka, India
| | - Vaibhav Pandita
- Department of Oral Medicine and Radiology, Nitte (Deemed to be University), AB Shetty Memorial Institute of Dental Sciences, Mangalore, Karnataka, India
| |
Collapse
|
616
|
Zhou N, Velez MA, Bachrach B, Gukasyan J, Fares CM, Cummings AL, Lind-Lebuffe JP, Akingbemi WO, Li DY, Brodrick PM, Yessuf NM, Rettinger S, Grogan T, Rochigneux P, Goldman JW, Garon EB, Lisberg A. Immune checkpoint inhibitor induced thyroid dysfunction is a frequent event post-treatment in NSCLC. Lung Cancer 2021; 161:34-41. [PMID: 34507111 PMCID: PMC8923054 DOI: 10.1016/j.lungcan.2021.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/27/2021] [Accepted: 08/20/2021] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Thyroid dysfunction is the most frequent endocrine immune related adverse event (irAE) in non-small cell lung cancer (NSCLC), typically arising 3-6 months into immune checkpoint inhibitor (ICI) therapy, but arising after ICI cessation, in some cases. Due to limited post-treatment adverse event reporting requirements on ICI trials, the incidence of ICI-induced thyroid dysfunction arising after therapy is unclear. We investigated ICI-induced thyroid dysfunction in a cohort of 294 NSCLC patients, with a specific focus on the post-treatment setting. METHODS Retrospective analysis of ICI-induced thyroid dysfunction (clinically acted upon or laboratory only) was performed in 294 UCLA NSCLC patients treated 2012-2018. Clinically acted upon thyroid dysfunction was defined as thyroid diagnosis documentation and/or thyroid medication administration. Laboratory only dysfunction was defined as abnormal thyroid labs in the absence of clinical action. Timing of thyroid dysfunction relative to ICI treatment and thyroid monitoring patterns were also assessed. RESULTS 82% (241/294) of ICI treated NSCLC patients had thyroid labs during treatment. Of these 241 patients, 13% (31/241) had clinically acted upon thyroid dysfunction prior to, 8% (18/241) during, and 4% (9/241) after ICI. Most patients, 66% (159/241), did not have thyroid labs after ICI, but in the 53 patients with labs and no prior clinical dysfunction, 17% (9/53) developed clinical dysfunction after ICI. In these 9 patients, median time from ICI initiation to dysfunction was 253 days. Two patients with post-treatment laboratory only dysfunction were observed. CONCLUSIONS ICI-induced thyroid dysfunction arising post-treatment appears more common than previously appreciated, warranting additional evaluation.
Collapse
Affiliation(s)
- Nanruoyi Zhou
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Maria A Velez
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Benjamin Bachrach
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Jaklin Gukasyan
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Charlene M Fares
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Amy L Cummings
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Jackson P Lind-Lebuffe
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Wisdom O Akingbemi
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Debory Y Li
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Paige M Brodrick
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Nawal M Yessuf
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Sarah Rettinger
- Department of Endocrinology, Providence Health & Services, Santa Monica, CA, USA
| | - Tristan Grogan
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Philippe Rochigneux
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA; Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Jonathan W Goldman
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Edward B Garon
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Aaron Lisberg
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
617
|
Segués A, van Duijnhoven SMJ, Parade M, Driessen L, Vukovic N, Zaiss D, Sijts AJAM, Berraondo P, van Elsas A. Generation and characterization of novel co-stimulatory anti-mouse TNFR2 antibodies. J Immunol Methods 2021; 499:113173. [PMID: 34699840 DOI: 10.1016/j.jim.2021.113173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 10/20/2022]
Abstract
Tumor necrosis factor receptor 2 (TNFR2) has gained much research interest in recent years because of its potential pivotal role in autoimmune disease and cancer. However, its function in regulating different immune cells is not well understood. There is a need for well-characterized reagents to selectively modulate TNFR2 function, thereby enabling definition of TNFR2-dependent biology in human and mouse surrogate models. Here, we describe the generation, production, purification, and characterization of a panel of novel antibodies targeting mouse TNFR2. The antibodies display functional differences in binding affinity and potency to block TNFα. Furthermore, epitope binding showed that the anti-mTNFR2 antibodies target different domains on the TNFR2 protein, associated with varying capacity to enhance CD8+ T-cell activation and costimulation. Moreover, the anti-TNFR2 antibodies demonstrate binding to isolated splenic mouse Tregs ex vivo and activated CD8+ cells, reinforcing their potential use to establish TNFR2-dependent immune modulation in translational models of autoimmunity and cancer.
Collapse
Affiliation(s)
- Aina Segués
- Aduro Biotech Europe, Oss, the Netherlands; Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands; Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, United Kingdom
| | | | | | | | - Nataša Vukovic
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, United Kingdom
| | - Dietmar Zaiss
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, United Kingdom; Institute of Immune Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Alice J A M Sijts
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Pedro Berraondo
- Division of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | | |
Collapse
|
618
|
Tseng PJ, Yan MT. Acute Diffuse Renal Tubulopathy in a Patient With Lung Cancer: A Case Report. Front Med (Lausanne) 2021; 8:742489. [PMID: 34671624 PMCID: PMC8520996 DOI: 10.3389/fmed.2021.742489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/03/2021] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoints inhibitors (ICPIs), as either a frontline or adjuvant therapy, showed favorable outcomes among diverse malignancies. Immune-related adverse events (IRAEs) are increasingly encountered, but the kidneys are rarely affected. A 67-year-old man with stage IV squamous cell carcinoma of the lung presented with acute kidney injury and hypercalcemia secondary to bone metastasis. After an aggressive saline infusion and subcutaneous denosumab 60mg administration, his renal function and serum calcium level were recovered on day 4. Due to his intolerance to chemotherapy, immunotherapy with a monoclonal antibody targeting programmed cell death protein-1 (PD-1), pembrolizumab 2mg/kg, was used on day 4. On day 11, polyuria, non-albumin dominant proteinuria, and severe deficiencies of electrolytes (potassium 2.5 mmol/L, calcium 5.5 mg/dL, magnesium 1.3 mg/dL, and phosphate 1.5 mg/dL) along with concomitant renal wasting were developed acutely. Except for postponing the next pembrolizumab, prednisolone at 1 mg/kg/day was given on day 13. On day 27, his polyuria subsided and urine protein loss resolved. Serum levels of potassium, phosphate, calcium, and magnesium all returned within the reference range. This case highlighted that renal IRAEs, even though uncommon, could be severe and potentially life-threatening if left unrecognized and untreated. Early recognition of renal IRAEs and prompt withdrawal of ICPIs may result in lower renal morbidity.
Collapse
Affiliation(s)
- Po-Jung Tseng
- Division of Nephrology, Department of Medicine, Cathay General Hospital, School of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| | - Ming-Tso Yan
- Division of Nephrology, Department of Medicine, Cathay General Hospital, School of Medicine, Fu-Jen Catholic University, Taipei, Taiwan.,National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei, Taiwan
| |
Collapse
|
619
|
Ma P, Jin X, Fan Z, Wang Z, Yue S, Wu C, Chen S, Wu Y, Chen M, Gu D, Zhang S, Mao R, Fan Y. Super-enhancer receives signals from the extracellular matrix to induce PD-L1-mediated immune evasion via integrin/BRAF/TAK1/ERK/ETV4 signaling. Cancer Biol Med 2021; 19:j.issn.2095-3941.2021.0137. [PMID: 34623791 PMCID: PMC9196059 DOI: 10.20892/j.issn.2095-3941.2021.0137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE PD-L1 and PD-L2 expression levels determine immune evasion and the therapeutic efficacy of immune checkpoint blockade. The factors that drive inducible PD-L1 expression have been extensively studied, but mechanisms that result in constitutive PD-L1 expression in cancer cells are largely unknown. METHODS DNA elements were deleted in cells by CRISPR/Cas9-mediated knockout. Protein function was inhibited by chemical inhibitors. Protein levels were examined by Western blot, mRNA levels were examined by real-time RT-PCR, and surface protein expression was determined by cellular immunofluorescence and flow cytometry. Immune evasion was examined by in vitro T cell-mediated killing. RESULTS We determined the core regions (chr9: 5, 496, 378-5, 499, 663) of a previously identified PD-L1L2-super-enhancer (SE). Through systematic analysis, we found that the E26 transformation-specific (ETS) variant transcription factor (ETV4) bound to this core DNA region but not to DNA surrounding PD-L1L2SE. Genetic knockout of ETV4 dramatically reduced the expressions of both PD-L1 and PD-L2. ETV4 transcription was dependent on ERK activation, and BRAF/TAK1-induced ERK activation was dependent on extracellular signaling from αvβ3 integrin, which profoundly affected ETV4 transcription and PD-L1/L2 expression. Genetic silencing or pharmacological inhibition of components of the PD-L1L2-SE-associated pathway rendered cancer cells susceptible to T cell-mediated killing. CONCLUSIONS We identified a pathway originating from the extracellular matrix that signaled via integrin/BRAF/TAK1/ERK/ETV4 to PD-L1L2-SE to induce PD-L1-mediated immune evasion. These results provided new insights into PD-L1L2-SE activation and pathways associated with immune checkpoint regulation in cancer.
Collapse
Affiliation(s)
- Panpan Ma
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
- Department of Clinical Laboratory, Yancheng No. 1 People's Hospital, Yancheng 224005, China
| | - Xinxin Jin
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Zhiwei Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Zhou Wang
- School of Life Sciences, Nantong University, Nantong 226001, China
| | - Suhui Yue
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Changyue Wu
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Shiyin Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Yuanyuan Wu
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Miaomiao Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Donghua Gu
- The Department of Urology, the Second Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Siliang Zhang
- The Department of Radiotherapy Oncology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| | - Yihui Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
620
|
Morelli T, Fujita K, Redelman-Sidi G, Elkington PT. Infections due to dysregulated immunity: an emerging complication of cancer immunotherapy. Thorax 2021; 77:304-311. [PMID: 34607905 PMCID: PMC8867274 DOI: 10.1136/thoraxjnl-2021-217260] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/12/2021] [Indexed: 01/01/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionised cancer treatment. However, immune-related adverse events (irAEs) are a common side effect which can mimic infection. Additionally, treatment of irAEs with corticosteroids and other immunosuppressant agents can lead to opportunistic infection, which we have classed as immunotherapy infections due to immunosuppression. However, emerging reports demonstrate that some infections can be precipitated by ICIs in the absence of immunosuppressive treatment, in contrast to the majority of reported cases. These infections are characterised by a dysregulated inflammatory immune response, and so we propose they are described as immunotherapy infections due to dysregulated immunity. This review summarises the rapidly emerging evidence of these phenomena and proposes a new framework for considering infection in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Tommaso Morelli
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Kohei Fujita
- Respiratory Medicine, National Hospital Organisation Kyoto Medical Center, Kyoto, Japan
| | - Gil Redelman-Sidi
- Division of Infectious Diseases, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Paul T Elkington
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK .,Institute for Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
621
|
Igarashi K, Cabral H, Hong T, Anraku Y, Mpekris F, Stylianopoulos T, Khan T, Matsumoto A, Kataoka K, Matsumoto Y, Yamasoba T. Vascular Bursts Act as a Versatile Tumor Vessel Permeation Route for Blood-Borne Particles and Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103751. [PMID: 34528759 DOI: 10.1002/smll.202103751] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Indexed: 06/13/2023]
Abstract
Dynamic bursting in tumor vasculature has recently sparked interest as a novel particle transportation route for drug delivery. These bursts facilitate the transport of sub-100 nm nanoparticles into tumors, though their contribution on the access of other blood-borne particles remains unknown. To evaluate the versatility of this phenomenon, the in vivo kinetics of a variety of intravenously injected particles and their penetration in tumor xenografts and allografts are compared. Dextran, polymeric micelles, liposomes, and polymeric vesicles with diameters ranging from 32 to 302 nm are found to colocalize in virtually all vascular bursts. By mathematical modeling, the burst vent size is estimated to be 625 nm or larger, indicating the dynamic and stochastic formation of large permeation routes in tumor vasculature. Furthermore, some burst vents are found to be µm-sized, allowing the transport of 1 µm microspheres. Moreover, antibody drugs and platelets are capable of utilizing vascular burst transportation, demonstrating the application of this phenomenon to other types of therapeutics and cellular components. These findings indicate the vast potential of vascular bursts, extending the biological and therapeutic significance of this phenomenon to a wide range of blood-borne particles and cells.
Collapse
Affiliation(s)
- Kazunori Igarashi
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Taehun Hong
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yasutaka Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, 1678, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, 1678, Cyprus
| | - Thahomina Khan
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Akira Matsumoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
- Kanagawa Institute of Industrial Science and Technology, 3-25-13 Tono-machi, Kawasaki-ku, Kawasaki City, Kanagawa, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tono-machi, Kawasaki-ku, Kawasaki City, Kanagawa, 210-0821, Japan
- Institute of Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 133-0033, Japan
| | - Yu Matsumoto
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tatsuya Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
622
|
Ferreira CA, Heidari P, Ataeinia B, Sinevici N, Sise ME, Colvin RB, Wehrenberg-Klee E, Mahmood U. Non-invasive Detection of Immunotherapy-Induced Adverse Events. Clin Cancer Res 2021; 27:5353-5364. [PMID: 34253581 PMCID: PMC8752648 DOI: 10.1158/1078-0432.ccr-20-4641] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/27/2021] [Accepted: 07/08/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Cancer immunotherapy has markedly improved the prognosis of patients with a broad variety of malignancies. However, benefits are weighed against unique toxicities, with immune-related adverse events (irAE) that are frequent and potentially life-threatening. The diagnosis and management of these events are challenging due to heterogeneity of timing onset, multiplicity of affected organs, and lack of non-invasive monitoring techniques. We demonstrate the use of a granzyme B-targeted PET imaging agent (GZP) for irAE identification in a murine model. EXPERIMENTAL DESIGN We generated a model of immunotherapy-induced adverse events in Foxp3-DTR-GFP mice bearing MC38 tumors. GZP PET imaging was performed to evaluate organs non-invasively. We validated imaging with ex vivo analysis, correlating the establishment of these events with the presence of immune infiltrates and granzyme B upregulation in tissue. To demonstrate the clinical relevance of our findings, the presence of granzyme B was identified through immunofluorescence staining in tissue samples of patients with confirmed checkpoint inhibitor-associated adverse events. RESULTS GZP PET imaging revealed differential uptake in organs affected by irAEs, such as colon, spleen, and kidney, which significantly diminished after administration of the immunosuppressor dexamethasone. The presence of granzyme B and immune infiltrates were confirmed histologically and correlated with significantly higher uptake in PET imaging. The presence of granzyme B was also confirmed in samples from patients that presented with clinical irAEs. CONCLUSIONS We demonstrate an interconnection between the establishment of irAEs and granzyme B presence and, for the first time, the visualization of those events through PET imaging.
Collapse
Affiliation(s)
| | - Pedram Heidari
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Bahar Ataeinia
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Nicoleta Sinevici
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Meghan E. Sise
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts
| | - Robert B. Colvin
- Department of Pathology and Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Umar Mahmood
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
623
|
Jiang C, Zhang L, Xu X, Qi M, Zhang J, He S, Tian Q, Song S. Engineering a Smart Agent for Enhanced Immunotherapy Effect by Simultaneously Blocking PD-L1 and CTLA-4. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102500. [PMID: 34473430 PMCID: PMC8529437 DOI: 10.1002/advs.202102500] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/04/2021] [Indexed: 05/06/2023]
Abstract
Combinations of immune checkpoint therapies show encouraging results in the treatment of many human cancers. However, the higher costs and greater side effects of such combinations compared with single-agent immunotherapies limit their further applications. In this work, a novel smart agent, KN046@19 F-ZIF-8, is developed to overcome these limitations. KN046 is a novel recombinant humanized PD-L1/CTLA-4 bispecific single-domain antibody-Fc fusion protein, which can bind to both PD-L1 and CTLA-4 effectively. ZIF-8 is a smart delivery system, which can safely and effectively deliver KN406 to a tumor. In vitro and in vivo results demonstrate that the smart agent KN046@19 F-ZIF-8 not only improves the immune response rate of the antibody drug in treatment of tumors but also reduces its toxic side effects, thereby achieving excellent antitumor efficacy. This study provides an engineering strategy for clinical applications of a more effective immunotherapy.
Collapse
Affiliation(s)
- Chunjuan Jiang
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Le Zhang
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
- Department of Research and DevelopmentShanghai Proton and Heavy Ion CenterShanghai201321China
| | - Xiaoping Xu
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Ming Qi
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Jianping Zhang
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Simin He
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Qiwei Tian
- Shanghai Key Laboratory of Molecular ImagingShanghai University of Medicine and Health SciencesShanghai201318China
| | - Shaoli Song
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
- Department of Research and DevelopmentShanghai Proton and Heavy Ion CenterShanghai201321China
| |
Collapse
|
624
|
Lu J, Zhang Y, Lou Y, Yan B, Zou B, Hu M, Wang Y, Chen Y, Yang Z, Wang H, Zhang W, Han B. ctDNA-Profiling-Based UBL Biological Process Mutation Status as a Predictor of Atezolizumab Response Among TP53-Negative NSCLC Patients. Front Genet 2021; 12:723670. [PMID: 34557222 PMCID: PMC8452871 DOI: 10.3389/fgene.2021.723670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023] Open
Abstract
Atezolizumab, an immune checkpoint inhibitor, has been approved for use in clinical practice in non-small cell lung cancer (NSCLC) patients, but potential biomarkers for response stratification still need further screening. In the present study, a total of 399 patients with high-quality ctDNA profiling results were included. The mutation status of ubiquitin-like conjugation (UBL) biological process genes (including ABL1, APC, LRP6, FUBP1, KEAP1, and TOP2A) and clinical information were further integrated. The results suggested that the patients with the clinical characteristics of male or history of smoking had a higher frequency of UBL mutation positivity [UBL (+)]; the patients who were UBL (+) had shorter progression-free survival (PFS) (1.69 vs. 3.22 months, p = 0.0007) and overall survival (8.61 vs. 16.10 months, p < 0.0001) than those patients with UBL mutation negativity [UBL (–)]; and more promising predictive values were shown in the smoker subgroup and ≤ 3 metastasis subgroup. More interestingly, we found the predictor has more performance in TP53-negative cohorts [training in an independent POPLAR and OAK cohorts (n = 200), and validation in an independent MSKCC cohort (n = 127)]. Overall, this study provides a predictor, UBL biological process gene mutation status, not only for identifying NSCLC patients who may respond to atezolizumab therapy but also for screening out the potential NSCLC responders who received other immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Jun Lu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Translational Medical Research Platform for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanwei Zhang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqing Lou
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Yan
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Clinical Research Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Benkun Zou
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Minjuan Hu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanan Wang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ya Chen
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengyu Yang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huimin Wang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Translational Medical Research Platform for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
625
|
Paderi A, Gambale E, Botteri C, Giorgione R, Lavacchi D, Brugia M, Mazzoni F, Giommoni E, Bormioli S, Amedei A, Pillozzi S, Matucci Cerinic M, Antonuzzo L. Association of Systemic Steroid Treatment and Outcome in Patients Treated with Immune Checkpoint Inhibitors: A Real-World Analysis. Molecules 2021; 26:molecules26195789. [PMID: 34641331 PMCID: PMC8510096 DOI: 10.3390/molecules26195789] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 11/18/2022] Open
Abstract
Background: Immune-related adverse events (irAEs) are inflammatory side effects, which can occur during immune-checkpoint(s) inhibitors (ICIs) therapy. Steroids are the first-line agents to manage irAEs because of their immunosuppressive properties. However, it is still debated whether or when steroids can be administered without abrogating the therapeutic efforts of immunotherapy. Methods: We retrospectively evaluated 146 patients with metastatic non-small cell lung cancer (NSCLC), melanoma and renal cell carcinoma (RCC) treated with ICIs. We assessed the progression-free survival (PFS) of patients treated with steroids due to an irAE compared to a no-steroid group. Results: The early treatment with steroid (within the first 30 days from the beginning of immunotherapy) was not related to a shorter PFS (p = 0.077). Interestingly, patients who were treated with steroids after 30 days from the start of immunotherapy had significantly longer PFS (p = 0.017). In a multivariate analysis, treatment with steroids after 30 days was an independent prognostic factor for PFS (HR: 0.59 [95% CI 0.36–0.97], p = 0.037). Conclusions: This retrospective study points out that early systemic steroids administration to manage irAEs might not have a detrimental effect on patient clinical outcome in NSCLC, melanoma and RCC patients.
Collapse
Affiliation(s)
- Agnese Paderi
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Elisabetta Gambale
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Cristina Botteri
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Roberta Giorgione
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Daniele Lavacchi
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Marco Brugia
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Francesca Mazzoni
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Elisa Giommoni
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Susanna Bormioli
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.); (A.A.); (M.M.C.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.); (A.A.); (M.M.C.)
| | - Serena Pillozzi
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Marco Matucci Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.); (A.A.); (M.M.C.)
| | - Lorenzo Antonuzzo
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.); (A.A.); (M.M.C.)
- Correspondence:
| |
Collapse
|
626
|
Deng C, Yang M, Jiang H, Wang R, Yang Z, Sun H, Cui H. Immune-Related Multiple-Organs Injuries Following ICI Treatment With Tislelizumab in an Advanced Non-Small Cell Lung Cancer Patient: A Case Report. Front Oncol 2021; 11:664809. [PMID: 34540659 PMCID: PMC8443792 DOI: 10.3389/fonc.2021.664809] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 08/12/2021] [Indexed: 01/14/2023] Open
Abstract
Immune-related adverse events (irAEs) following treatment with immune checkpoint inhibitors (ICIs) can affect almost any organ systems. Multiple-organs irAEs are a rare occurrence which makes its management and treatment very challenging. This is a case report of a 71-year-old man with advanced non-small cell lung cancer (NSCLC) who developed multiple-organs irAEs (lung, muscle, myocardium, liver, and pituitary) after a single cycle (21 days) of the BGB-A317 (Tislelizumab). After more than two months of immunosuppression treatment with glucocorticoids, the tumor and inflammatory lesions in the lung were reduced. The levels of serum creatase, cardiac troponin T (TNT), and hepatic transaminase were also reduced. Four months after the termination of ICI therapy, the lung tumor reappeared in the previous site. This rare case report supplies several experiences in the management of multiple-organs irAEs, including full-scale monitoring of immunological indicators, early differential diagnosis, and prompt glucocorticoid therapy. This patient was not a candidate for the ICI re-challenge therapy due to the number and seriousness of irAEs. Multiple-organs irAEs add complexity to the management, and additional research is needed to develop optimal therapeutic guidelines.
Collapse
Affiliation(s)
- Chao Deng
- Department of Medical Oncology, Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Meng Yang
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Hong Jiang
- Department of Cardiology, Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Renbin Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Zhaojun Yang
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - Hongliang Sun
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Huijuan Cui
- Department of Medical Oncology, Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
627
|
Clinton JW, Kiparizoska S, Aggarwal S, Woo S, Davis W, Lewis JH. Drug-Induced Liver Injury: Highlights and Controversies in the Recent Literature. Drug Saf 2021; 44:1125-1149. [PMID: 34533782 PMCID: PMC8447115 DOI: 10.1007/s40264-021-01109-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 12/13/2022]
Abstract
Drug-induced liver injury (DILI) remains an important, yet challenging diagnosis for physicians. Each year, additional drugs are implicated in DILI and this year was no different, with more than 1400 articles published on the subject. This review examines some of the most significant highlights and controversies in DILI-related research over the past year and their implications for clinical practice. Several new drugs were approved by the US Food and Drug Administration including a number of drugs implicated in causing DILI, particularly among the chemotherapeutic classes. The COVID-19 pandemic was also a major focus of attention in 2020 and we discuss some of the notable aspects of COVID-19-related liver injury and its implications for diagnosing DILI. Updates in diagnostic and causality assessments related to DILI such as the Roussel Uclaf Causality Assessment Method are included, mindful that there is still no single biomarker or diagnostic tool to unequivocally diagnose DILI. Glutamate dehydrogenase received renewed attention as being more specific than alanine aminotransferase. There were a few new reports of previously unrecognized hepatotoxins, including immune modulators and novel gene therapy drugs that we highlight. Updates and new developments of previously described hepatotoxins, such as immune checkpoint inhibitors and anti-tuberculosis drugs are reviewed. Finally, novel technologies such as organoid culture systems to better predict DILI preclinically may be coming of age and determinants of hepatocyte loss, such as calculating PALT are poised to improve our current means of estimating DILI severity and the risk of acute liver failure.
Collapse
Affiliation(s)
- Joseph William Clinton
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA.
| | - Sara Kiparizoska
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Soorya Aggarwal
- Division of Gastroenterology and Hepatology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Stephanie Woo
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - William Davis
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - James H Lewis
- Division of Gastroenterology and Hepatology, Medstar Georgetown University Hospital, Washington, DC, USA
| |
Collapse
|
628
|
Zhou L, Wei X. Ocular Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitors in Lung Cancer. Front Immunol 2021; 12:701951. [PMID: 34504488 PMCID: PMC8421677 DOI: 10.3389/fimmu.2021.701951] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/05/2021] [Indexed: 02/05/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are novel immunotherapy-based drugs that have become increasingly popular in the treatment of lung cancer. Researchers have recognized ocular immune-related adverse events (irAEs) secondary to ICIs because of their vision-threatening characteristics. However, they are incompletely characterized and no studies have reported the ICI-related ocular irAEs in lung cancer. Therefore, we aimed to comprehensively illustrate the clinical characteristics, contributory factors, diagnosis, and management of ICI-related ocular irAEs in lung cancer, based on previously reported 79 patients. Ophthalmoplegia (40.51%), uveitis (20.25%), and dry eye (17.72%) were the most common ICI-related ocular irAEs in lung cancer. Ptosis was the most common (36.71%) and the highest mortality (23.33%) of ophthalmoplegia. Patients in Asia and patients who underwent combination therapy with programmed cell death-1 and cytotoxic T-lymphocyte-associated antigen 4 inhibitors demonstrated significantly higher frequency of ophthalmoplegia than other ocular irAEs. Most ICI-related ophthalmoplegia and uveitis in lung cancer were observed in the first 10 weeks following the initiation of ICIs. Furthermore, the onset time of dry eye and other ocular irAEs was much longer. In addition, 92.31% of the patients with ocular irAEs other than ophthalmoplegia could be remised. In conclusion, ocular irAEs secondary to ICIs in lung cancer are non-negligible, particularly ophthalmoplegia. Ethnicity and the type of ICIs play important roles in the distribution of ocular irAEs. ICI-related ophthalmoplegia in lung cancer presented with early onset and worse prognosis features, thus necessitating further attention.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
629
|
Zhang PP, Wang J, Ding DZ, Zhang L, Cheng C, Chen DK. Efficacy and safety of PD-1/PD-L1 inhibitors combined with CTLA-4 inhibitor versus chemotherapy for advanced lung cancer: A meta-analysis. Medicine (Baltimore) 2021; 100:e27121. [PMID: 34477154 PMCID: PMC8416009 DOI: 10.1097/md.0000000000027121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND This meta-analysis was performed to compare efficacy and tolerability between antiprogrammed cell death (PD-1)/programmed cell death-ligand-1 (PD-L1) + anticytotoxic T-lymphocyte-associated protein-4 (CTLA-4) treatment and chemotherapy in advanced lung cancer. METHODS Cochrane Library, Embase, and PubMed databases were searched for potential articles. The fixed-effect model or random-effect model was adopted for pooled analysis based on the I2 and P-value. RESULTS Six articles with 1338 patients were identified and subjected to meta-analysis. Compared with chemotherapy, anti-PD-1/PD-L1 + anti-CTLA-4 treatment could significantly improve the overall survival (hazard ratio [HR] = 0.78, 95%confidence interval [CI]: 0.71-0.84, P = .21) and progression-free survival (HR = 0.77, 95%CI: 0.71-0.83, P = .30) of advanced lung cancer patients. Moreover, there was no obvious difference in the incidence of 3 to 4 adverse events (AEs) serious adverse reactions (HR = 1.35, 95%CI: 0.66-2.74, P < .00001) between the 2 treatment groups, but the incidence rates of AEs leading to discontinuation (HR = 2.56, 95%CI: 1.53-4.30, P < .00001) and AEs leading to death (HR = 2.10, 95%CI: 1.21-3.63, P = .20) were higher. Furthermore, no remarkable differences in objective response rate (HR = 1.31, 95%CI: 0.97-1.77, P = .02) were observed between the 2 groups. CONCLUSION Our meta-analysis revealed that PD-1/PD-L1 inhibitors plus CTLA-4 inhibitor could markedly improve the endpoint outcomes of patients compared with chemotherapy alone, and did not significantly increase the serious adverse reactions. Thus, it can serve as a new treatment strategy for advanced lung cancer.
Collapse
Affiliation(s)
- Pei-Pei Zhang
- Department of Oncology, Affiliated Hospital of Nantong University, Tongzhou District People's Hospital, Nantong City, Jiangsu Province, No. 115, Jianshe Road, Jinsha Town, Tongzhou District, Nantong City, Jiangsu Province, China
| | - Juan Wang
- Department of Oncology, Affiliated Hospital of Nantong University, Tongzhou District People's Hospital, Nantong City, Jiangsu Province, No. 115, Jianshe Road, Jinsha Town, Tongzhou District, Nantong City, Jiangsu Province, China
| | - Da-Zhi Ding
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Tongzhou District People's Hospital, Nantong City, Jiangsu Province, No. 115, Jianshe Road, Jinsha Town, Tongzhou District, Nantong City, Jiangsu Province, China
| | - Li Zhang
- Department of Oncology, Affiliated Hospital of Nantong University, Tongzhou District People's Hospital, Nantong City, Jiangsu Province, No. 115, Jianshe Road, Jinsha Town, Tongzhou District, Nantong City, Jiangsu Province, China
| | - Chun Cheng
- Department of Immunology, School of Medicine, Nantong University, Jiangsu Province, No. 19, Qixiu Road, Chongchuan District, Nantong City, Jiangsu Province, China
| | - Da-Ke Chen
- Department of Oncology, Affiliated Hospital of Nantong University, Tongzhou District People's Hospital, Nantong City, Jiangsu Province, No. 115, Jianshe Road, Jinsha Town, Tongzhou District, Nantong City, Jiangsu Province, China
| |
Collapse
|
630
|
Zubiri L, Molina GE, Mooradian MJ, Cohen J, Durbin SM, Petrillo L, Boland GM, Juric D, Dougan M, Thomas MF, Faje AT, Rengarajan M, Guidon AC, Chen ST, Okin D, Medoff BD, Nasrallah M, Kohler MJ, Schoenfeld SR, Karp-Leaf RS, Sise ME, Neilan TG, Zlotoff DA, Farmer JR, Bardia A, Sullivan RJ, Blum SM, Semenov YR, Villani AC, Reynolds KL. Effect of a multidisciplinary Severe Immunotherapy Complications Service on outcomes for patients receiving immune checkpoint inhibitor therapy for cancer. J Immunother Cancer 2021; 9:e002886. [PMID: 34544895 PMCID: PMC8454442 DOI: 10.1136/jitc-2021-002886] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In 2017, Massachusetts General Hospital implemented the Severe Immunotherapy Complications (SIC) Service, a multidisciplinary care team for patients hospitalized with immune-related adverse events (irAEs), a unique spectrum of toxicities associated with immune checkpoint inhibitors (ICIs). This study's objectives were to evaluate the intervention's (1) effect on patient outcomes and healthcare utilization, and (2) ability to collect biological samples via a central infrastructure, in order to study the mechanisms responsible for irAEs. METHODS A hospital database was used to identify patients who received ICIs for a malignancy and were hospitalized with severe irAEs, before (April 2, 2016-October 3, 2017) and after (October 3, 2017-October 24, 2018) SIC Service initiation. The primary outcome was readmission rate after index hospitalization. Secondary outcomes included length of stay (LOS) for admissions, corticosteroid and non-steroidal second-line immunosuppression use, ICI discontinuation, and inpatient mortality. RESULTS In the pre-SIC period, 127 of 1169 patients treated with ICIs were hospitalized for irAEs; in the post-SIC period, 122 of 1159. After SIC service initiation, reductions were observed in irAE readmission rate (14.8% post-SIC vs 25.9% pre-SIC; OR 0.46; 95% CI 0.22 to 0.95; p=0.036) and readmission LOS (median 6 days post-SIC vs 7 days pre-SIC; 95% CI -16.03 to -0.14; p=0.046). No significant pre-initiation and post-initiation differences were detected in corticosteroid use, second-line immunosuppression, ICI discontinuation, or inpatient mortality rates. The SIC Service collected 789 blood and tissue samples from 234 patients with suspected irAEs. CONCLUSIONS This is the first study to report that establishing a highly subspecialized care team focused on irAEs is associated with improved patient outcomes and reduced healthcare utilization. Furthermore, the SIC Service successfully integrated blood and tissue collection safety into routine care.
Collapse
Affiliation(s)
- Leyre Zubiri
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gabriel E Molina
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meghan J Mooradian
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Justine Cohen
- Division of Oncology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Sienna M Durbin
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura Petrillo
- Division of Palliative Care, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Genevieve M Boland
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dejan Juric
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Dougan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Molly F Thomas
- Division of Gastroenterology, Department of Medicine, Mass General Center for Cancer Research, Division of Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alex T Faje
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michelle Rengarajan
- Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amanda C Guidon
- Division of Neuromuscular Disorders, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven T Chen
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Okin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin D Medoff
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mazen Nasrallah
- Division of Rheumatology, Allergy, Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Minna J Kohler
- Division of Rheumatology, Allergy, Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara R Schoenfeld
- Division of Rheumatology, Allergy, Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca S Karp-Leaf
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meghan E Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel A Zlotoff
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jocelyn R Farmer
- Division of Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aditya Bardia
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan J Sullivan
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven M Blum
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yevgeniy R Semenov
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandra-Chloé Villani
- Massachusetts General Hospital Center for Immunology and Inflammatory Diseases, Mass General Center for Cancer Research, Division of Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kerry L Reynolds
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
631
|
Kitchlu A, Jhaveri KD, Sprangers B, Yanagita M, Wanchoo R. Immune checkpoint inhibitor use in patients with end-stage kidney disease: an analysis of reported cases and literature review. Clin Kidney J 2021; 14:2012-2022. [PMID: 34476087 PMCID: PMC8406068 DOI: 10.1093/ckj/sfab090] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 01/13/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs), immunomodulatory antibodies that are used to enhance the immune system, have substantially improved the prognosis of patients with advanced malignancy. As the use of ICI therapy becomes increasingly widespread across different types of cancer, their use in patients receiving dialysis is likely to increase. In this review we summarize the current literature on the use of ICIs in end-stage kidney disease (ESKD) patients and provide aggregate data from reported cases and series. Based on available pharmacological information, ICIs require no dosing adjustment in ESKD patients. Analysis of the reported cases in the literature demonstrates a similar incidence of immune-related adverse events in patients with ESKD receiving dialysis as compared with the general population (49%). Severe reactions graded as 3 and 4 have been seen in 15 patients (16%). As such, it is important that these patients are monitored very closely for immune-related adverse events; however, the risk of these adverse events should not preclude patients on dialysis from receiving these therapies. Cancer remission (complete and partial) was seen in close to 30% of patients, stable disease was seen in 28% and progression of disease in ∼36%. One-third of the patients died. Urothelial and renal cell cancer represented approximately half of all treated cancers and accounted for ∼50% of all deaths reported. Additional data in the dialysis population with the use of ICIs and involvement in prospective studies are needed to better assess outcomes, particularly within specific cancer types.
Collapse
Affiliation(s)
- Abhijat Kitchlu
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine, Northwell Health, Great Neck, NY, USA
| | - Ben Sprangers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Leuven, Belgium
- Division of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rimda Wanchoo
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine, Northwell Health, Great Neck, NY, USA
| |
Collapse
|
632
|
Russano M, Cortellini A, Giusti R, Russo A, Zoratto F, Rastelli F, Gelibter A, Chiari R, Nigro O, De Tursi M, Bracarda S, Gori S, Grossi F, Bersanelli M, Calvetti L, Di Noia V, Scartozzi M, Di Maio M, Bossi P, Falcone A, Citarella F, Pantano F, Ficorella C, Filetti M, Adamo V, Veltri E, Pergolesi F, Occhipinti MA, Nicolardi L, Tuzi A, Di Marino P, Macrini S, Inno A, Ghidini M, Buti S, Aprile G, Lai E, Audisio M, Intagliata S, Marconcini R, Brocco D, Porzio G, Piras M, Rijavec E, Simionato F, Natoli C, Tiseo M, Vincenzi B, Tonini G, Santini D. Clinical outcomes of NSCLC patients experiencing early immune-related adverse events to PD-1/PD-L1 checkpoint inhibitors leading to treatment discontinuation. Cancer Immunol Immunother 2021; 71:865-874. [PMID: 34462870 DOI: 10.1007/s00262-021-03045-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND The prognostic relevance of early immune-related adverse events (irAEs) in patients affected by non-small cell lung cancer (NSCLC) upon immunotherapy is not fully understood. METHODS The Leading to Treatment Discontinuation cohort included 24 patients experiencing severe irAEs after one of two administrations of single anti-PD-1/PD-L1 in any line setting for metastatic NSCLC between November 2015 and June 2019. The control cohort was composed of 526 patients treated with single anti-PD-1/PD-L1 in any line setting with no severe irAE reported. The primary end points were median progression-free survival, overall survival, objective response rate, risk of progression of disease and risk of death. The correlation of clinic pathological features with early severe irAEs represented the secondary end point. RESULTS Median PFS was 9.3 and 8.4 months, median OS was 12.0 months and 14.2 months at a median follow-up of 18.1 and 22.6 months in the LTD cohort and in the control cohort, respectively. The ORR was 40% (95% CI 17.2-78.8) in the LTD cohort and 32.7% (95% CI 27.8-38.2) in the control cohort. The risk of disease progression was higher in the LTD cohort (HR 2.52 [95% 1.10-5.78], P = .0288). CONCLUSIONS We found no survival benefit in LTD cohort compared to the control cohort. However, early and severe irAEs might underly an immune anti-tumor activation. We identified a significant association with first-line immune checkpoints inhibitors treatment and good PS. Further studies on risk prediction and management of serious and early irAEs in NSCLC patients are needed.
Collapse
Affiliation(s)
- Marco Russano
- Department of Medical Oncology, Campus Bio-Medico University, Via Alvaro del Portillo 200, 00128, Rome, Italy
| | - Alessio Cortellini
- Department of Biotechnology and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Alessandro Russo
- Medical Oncology, A.O. Papardo and Department of Human Pathology, University of Messina, Messina, Italy
| | | | | | - Alain Gelibter
- Medical Oncology (B), Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Rita Chiari
- Medical Oncology, Ospedali Riuniti Padova Sud "Madre Teresa Di Calcutta", Monselice, Italy
| | - Olga Nigro
- Medical Oncology, ASST-Sette Laghi, Varese, Italy
| | | | - Sergio Bracarda
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100, Terni, Italy
| | - Stefania Gori
- Oncology Department, IRCCS Sacro Cuore Don Calabria Hospital, 37024, Negrar, Verona, Italy
| | - Francesco Grossi
- Medical Oncology Unit, IRCCS Foundation Ca' Granda Maggiore Hospital Policlinic, Milan, Italy
| | - Melissa Bersanelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Lorenzo Calvetti
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| | | | - Mario Scartozzi
- Department of Medical Oncology, University Hospital of Cagliari, Cagliari, Italy
| | - Massimo Di Maio
- Department of Oncology, Medical Oncology Unit, Ordine Mauriziano Hospital, University of Turin, Turin, Italy
| | - Paolo Bossi
- Medical Oncology, ASST-Spedali Civili, University of Brescia, Brescia, Italy
| | - Alfredo Falcone
- Medical Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Fabrizio Citarella
- Department of Medical Oncology, Campus Bio-Medico University, Via Alvaro del Portillo 200, 00128, Rome, Italy.
| | - Francesco Pantano
- Department of Medical Oncology, Campus Bio-Medico University, Via Alvaro del Portillo 200, 00128, Rome, Italy
| | - Corrado Ficorella
- Department of Biotechnology and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Vincenzo Adamo
- Medical Oncology, A.O. Papardo and Department of Human Pathology, University of Messina, Messina, Italy
| | - Enzo Veltri
- Medical Oncology, Santa Maria Goretti Hospital, Latina, Italy
| | | | | | - Linda Nicolardi
- Medical Oncology, Ospedali Riuniti Padova Sud "Madre Teresa Di Calcutta", Monselice, Italy
| | | | | | - Serena Macrini
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100, Terni, Italy
| | - Alessandro Inno
- Oncology Department, IRCCS Sacro Cuore Don Calabria Hospital, 37024, Negrar, Verona, Italy
| | - Michele Ghidini
- Medical Oncology Unit, IRCCS Foundation Ca' Granda Maggiore Hospital Policlinic, Milan, Italy
| | - Sebastiano Buti
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| | - Eleonora Lai
- Department of Medical Oncology, University Hospital of Cagliari, Cagliari, Italy
| | - Marco Audisio
- Department of Oncology, Medical Oncology Unit, Ordine Mauriziano Hospital, University of Turin, Turin, Italy
| | | | | | - Davide Brocco
- Department of Pharmacy, G. d'Annunzio" University of Chieti-Pescara, Via Dei Vestini 31, 66100, Chieti, Italy
| | - Giampiero Porzio
- Department of Biotechnology and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marta Piras
- Medical Oncology, St. Andrea Hospital, Rome, Italy
| | - Erika Rijavec
- Medical Oncology Unit, IRCCS Foundation Ca' Granda Maggiore Hospital Policlinic, Milan, Italy
| | | | - Clara Natoli
- Clinical Oncology Unit, S.S. Annunziata Hospital, Chieti, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University, Via Alvaro del Portillo 200, 00128, Rome, Italy
| | - Giuseppe Tonini
- Department of Medical Oncology, Campus Bio-Medico University, Via Alvaro del Portillo 200, 00128, Rome, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University, Via Alvaro del Portillo 200, 00128, Rome, Italy
| |
Collapse
|
633
|
Martínez-Cortés F, Servín-Blanco R, Domínguez-Romero AN, Munguía ME, Guzman Valle J, Odales J, Gevorkian G, Manoutcharian K. Generation of cancer vaccine immunogens derived from Oncofetal antigen (OFA/iLRP) using variable epitope libraries tested in an aggressive breast cancer model. Mol Immunol 2021; 139:65-75. [PMID: 34454186 DOI: 10.1016/j.molimm.2021.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/30/2022]
Abstract
After decades of cancer vaccine efforts, there is an imperious necessity for novel ideas that may result in better tumor control in patients. We have proposed the use of a novel Variable Epitope Library (VEL) vaccine strategy, which incorporates an unprecedented number of mutated epitopes to target antigenic variability and break tolerance against tumor-associated antigens. Here, we used an oncofetal antigen/immature laminin receptor protein-derived sequence to generate 9-mer and 43-mer VEL immunogens. 4T1 tumor-bearing mice developed epitope-specific CD8+IFN-γ+ and CD4+IFN-γ+ T cell responses after treatment. Tumor and lung analysis demonstrated that VELs could increase the number of tumor-infiltrating lymphocytes with diverse effector functions while reducing the number of immunosuppressive myeloid-derived suppressor and regulatory T cells. Most importantly, VEL immunogens inhibited tumor growth and metastasis after a single dose. The results presented here are consistent with our previous studies and provide evidence for VEL immunogens' feasibility as promising cancer immunotherapy.
Collapse
Affiliation(s)
- Fernando Martínez-Cortés
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF, 04510, Mexico
| | - Rodolfo Servín-Blanco
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF, 04510, Mexico
| | - Allan Noé Domínguez-Romero
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF, 04510, Mexico
| | - María Elena Munguía
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF, 04510, Mexico
| | - Jesus Guzman Valle
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF, 04510, Mexico
| | - Josué Odales
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF, 04510, Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF, 04510, Mexico
| | - Karen Manoutcharian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF, 04510, Mexico.
| |
Collapse
|
634
|
Chanson N, Ramos-Casals M, Pundole X, Suijkerbuik K, José de Barros E Silva M, Lidar M, Benesova K, Leipe J, Acar-Denizli N, Pradère P, Michot JM, Voisin AL, Suárez-Almazor ME, Radstake TRD, Fernandes Moça Trevisani V, Schulze-Koops H, Melin A, Robert C, Mariette X, Baughman RP, Lambotte O. Immune checkpoint inhibitor-associated sarcoidosis: A usually benign disease that does not require immunotherapy discontinuation. Eur J Cancer 2021; 158:208-216. [PMID: 34452793 DOI: 10.1016/j.ejca.2021.05.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/27/2021] [Accepted: 05/24/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To analyse the clinical patterns of sarcoidosis triggered by immune checkpoint inhibitors (ICIs) in patients with cancer. PATIENTS AND METHODS The ImmunoCancer International Registry is a big data-sharing multidisciplinary network from 18 countries dedicated to evaluating the clinical research of immune-related adverse events related to cancer immunotherapies. RESULTS We identified 32 patients with biopsy-proven sarcoidosis. Underlying cancer included mainly melanoma (n = 24). Cancer immunotherapy consisted of monotherapy in 19 cases (anti-PD-1 in 18 and ipilimumab in 1) or combined ipilimumab + nivolumab in 13. The time median interval between initiation of ICI and sarcoidosis diagnosis was 3 months (range, 2-29 months). The use of combined ICI was associated with a shorter delay in developing sarcoidosis symptoms. The disease was symptomatic in 19 (59%) cases with mostly cutaneous, respiratory and general symptoms. The organs involved included mainly the mediastinal lymph nodes (n = 32), the lungs (n = 11), the skin (n = 10) and the eyes (n = 5). Pulmonary computed tomography studies showed bilateral hilar lymphadenopathy in all cases. There was no severe manifestation. Specific systemic therapy was required in only 12 patients (37%): oral glucocorticoids in 9, and hydroxychloroquine in 3. ICIs were held in 25 patients (78%) and definitively discontinued in 18 (56%) patients. Seven patients continued ICI treatment with a second flare in one case. In six additional patients, an ICI was reintroduced with no harm, and sarcoidosis relapsed in one of them. CONCLUSION Our study shows that ICI-related sarcoidosis seems to have a specific profile, possibly more benign than that of idiopathic sarcoidosis, and does not necessarily imply ICI discontinuation.
Collapse
Affiliation(s)
- Noémie Chanson
- AP-HP.Université Paris-Saclay, Hôpital Bicêtre, Department of Internal Medicine and Clinical Immunology, Le Kremlin Bicêtre, France; Université Paris-Saclay; INSERM; CEA, Centre Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Le Kremlin-Bicêtre, France
| | - Manuel Ramos-Casals
- Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain; Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain.
| | - Xerxes Pundole
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karijn Suijkerbuik
- Department of Medical Oncology, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | | | - Merav Lidar
- Rheumatology Unit, Sheba Medical Center, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Karolina Benesova
- Department of Internal Medicin and Rheumatology, Universitätsklinikum Heidelberg, Germany
| | - Jan Leipe
- Division of Rheumatology, Department of Medicine V, University Hospital Mannheim, Medical Faculty Mannheim of the University Heidelberg, German
| | - Nihan Acar-Denizli
- Department of Statistics, Faculty of Science and Letters, Mimar Sinan Fine Arts University, Istanbul, Turkey
| | - Pauline Pradère
- Department of Thoracic Surgery, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France
| | - Jean-Marie Michot
- Département D'Innovation Thérapeutique et D'Essais Précoces, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - Anne-Laure Voisin
- Unité Fonctionnelle de Pharmacovigilance, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - Maria E Suárez-Almazor
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy R D Radstake
- Department of Medical Oncology, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | | | - Hendrik Schulze-Koops
- Department of Internal Medicin and Rheumatology, Universitätsklinikum Heidelberg, Germany
| | - Audrey Melin
- Department of Dermatology, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - Caroline Robert
- Department of Dermatology, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - Xavier Mariette
- Université Paris-Saclay; INSERM; CEA, Centre Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Le Kremlin-Bicêtre, France; AP-HP.Université Paris-Saclay, Department of Rheumatology, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Robert P Baughman
- Department of Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Olivier Lambotte
- AP-HP.Université Paris-Saclay, Hôpital Bicêtre, Department of Internal Medicine and Clinical Immunology, Le Kremlin Bicêtre, France; Université Paris-Saclay; INSERM; CEA, Centre Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Le Kremlin-Bicêtre, France.
| | | | | | | | | | | | | |
Collapse
|
635
|
Gao S, Yang X, Xu J, Qiu N, Zhai G. Nanotechnology for Boosting Cancer Immunotherapy and Remodeling Tumor Microenvironment: The Horizons in Cancer Treatment. ACS NANO 2021; 15:12567-12603. [PMID: 34339170 DOI: 10.1021/acsnano.1c02103] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Immunotherapy that harnesses the human immune system to fight cancer has received widespread attention and become a mainstream strategy for cancer treatment. Cancer immunotherapy not only eliminates primary tumors but also treats metastasis and recurrence, representing a major advantage over traditional cancer treatments. Recently with the development of nanotechnology, there exists much work applying nanomaterials to cancer immunotherapy on the basis of their excellent physiochemical properties, such as efficient tissue-specific delivery function, huge specific surface area, and controllable surface chemistry. Consequently, nanotechnology holds significant potential in improving the efficacy of cancer immunotherapy. Nanotechnology-based immunotherapy mainly manifests its inhibitory effect on tumors via two different approaches: one is to produce an effective anti-tumor immune response during tumorigenesis, and the other is to enhance tumor immune defense ability by modulating the immune suppression mechanism in the tumor microenvironment. With the success of tumor immunotherapy, understanding the interaction between the immune system and smart nanomedicine has provided vigorous vitality for the development of cancer treatment. This review highlights the application, progress, and prospect of nanomedicine in the process of tumor immunoediting and also discusses several engineering methods to improve the efficiency of tumor treatment.
Collapse
Affiliation(s)
- Shan Gao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Xiaoye Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Jiangkang Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Na Qiu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Guangxi Zhai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| |
Collapse
|
636
|
Li J, Xie J, Wu D, Chen L, Gong Z, Wu R, Hu Y, Zhao J, Xu Y. A pan-cancer analysis revealed the role of the SLC16 family in cancer. Channels (Austin) 2021; 15:528-540. [PMID: 34424811 PMCID: PMC8386723 DOI: 10.1080/19336950.2021.1965422] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Cancer is one of the serious diseases that endanger human health and bring a heavy burden to world economic development. Although the current targeted therapy and immunotherapy have achieved initial results, the emergence of drug resistance shows that the existing research is far from enough. In recent years, the tumor microenvironment has been found to be an important condition for tumor development and has profound research value. The SLC16 family is a group of monocarboxylic acid transporters involved in cancer metabolism and the formation of the tumor microenvironment. However, there have been no generalized cancer studies in the SLC16 family. In this study, we conducted a pan-cancer analysis of the SLC16 family. The results showed that multiple members of the SLC16 family could be used as prognostic indicators for many tumors, and were associated with immune invasion and tumor stem cells. Therefore, the SLC16 family has extensive exploration value in the future.
Collapse
Affiliation(s)
- Jun Li
- Department of Thoracic Surgery, The First Affiliated Hospital to Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaheng Xie
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dan Wu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Liang Chen
- Department of General Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui, China
| | - Zetian Gong
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Wu
- Department of Digestive Endoscopy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yiming Hu
- College of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Jiangning Zhao
- Department of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yetao Xu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
637
|
He S, Li J, Cheng P, Zeng Z, Zhang C, Duan H, Pu K. Charge‐Reversal Polymer Nano‐modulators for Photodynamic Immunotherapy of Cancer. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202106392] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Shasha He
- School of Chemical and Biomedical Engineering School of Physical and Mathematical Sciences Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Jingchao Li
- School of Chemical and Biomedical Engineering School of Physical and Mathematical Sciences Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Penghui Cheng
- School of Chemical and Biomedical Engineering School of Physical and Mathematical Sciences Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Ziling Zeng
- School of Chemical and Biomedical Engineering School of Physical and Mathematical Sciences Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Chi Zhang
- School of Chemical and Biomedical Engineering School of Physical and Mathematical Sciences Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering School of Physical and Mathematical Sciences Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering School of Physical and Mathematical Sciences Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| |
Collapse
|
638
|
Campochiaro C, De Luca G, Dagna L. Cardiac immune-related adverse events: an immune-cardio-oncology puzzle. Eur J Heart Fail 2021; 23:1748-1749. [PMID: 34383998 DOI: 10.1002/ejhf.2329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 08/08/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
639
|
Kerns SJ, Belgur C, Petropolis D, Kanellias M, Barrile R, Sam J, Weinzierl T, Fauti T, Freimoser-Grundschober A, Eckmann J, Hage C, Geiger M, Ng PR, Tien-Street W, Manatakis DV, Micallef V, Gerard R, Bscheider M, Breous-Nystrom E, Schneider A, Giusti AM, Bertinetti-Lapatki C, Grant HS, Roth AB, Hamilton GA, Singer T, Karalis K, Moisan A, Bruenker P, Klein C, Bacac M, Gjorevski N, Cabon L. Human immunocompetent Organ-on-Chip platforms allow safety profiling of tumor-targeted T-cell bispecific antibodies. eLife 2021; 10:e67106. [PMID: 34378534 PMCID: PMC8373379 DOI: 10.7554/elife.67106] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022] Open
Abstract
Traditional drug safety assessment often fails to predict complications in humans, especially when the drug targets the immune system. Here, we show the unprecedented capability of two human Organs-on-Chips to evaluate the safety profile of T-cell bispecific antibodies (TCBs) targeting tumor antigens. Although promising for cancer immunotherapy, TCBs are associated with an on-target, off-tumor risk due to low levels of expression of tumor antigens in healthy tissues. We leveraged in vivo target expression and toxicity data of TCBs targeting folate receptor 1 (FOLR1) or carcinoembryonic antigen (CEA) to design and validate human immunocompetent Organs-on-Chips safety platforms. We discovered that the Lung-Chip and Intestine-Chip could reproduce and predict target-dependent TCB safety liabilities, based on sensitivity to key determinants thereof, such as target expression and antibody affinity. These novel tools broaden the research options available for mechanistic understandings of engineered therapeutic antibodies and assessing safety in tissues susceptible to adverse events.
Collapse
Affiliation(s)
| | | | | | | | - Riccardo Barrile
- Emulate IncBostonUnited States
- Department of Biomedical Engineering, University of CincinnatiCincinnatiUnited States
| | - Johannes Sam
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | - Tina Weinzierl
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | - Tanja Fauti
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | | | - Jan Eckmann
- Roche Pharma Research & Early Development, Roche Innovation Center MunichPenzbergGermany
| | - Carina Hage
- Roche Pharma Research & Early Development, Roche Innovation Center MunichPenzbergGermany
| | - Martina Geiger
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | | | | | | | - Virginie Micallef
- Roche Pharma Research & Early Development, Roche Innovation Center BaselBaselSwitzerland
| | - Regine Gerard
- Roche Pharma Research & Early Development, Roche Innovation Center BaselBaselSwitzerland
| | - Michael Bscheider
- Roche Pharma Research & Early Development, Roche Innovation Center BaselBaselSwitzerland
| | | | - Anneliese Schneider
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | - Anna Maria Giusti
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | | | | | - Adrian B Roth
- Roche Pharma Research & Early Development, Roche Innovation Center BaselBaselSwitzerland
| | | | - Thomas Singer
- Roche Pharma Research & Early Development, Roche Innovation Center BaselBaselSwitzerland
| | | | - Annie Moisan
- Roche Pharma Research & Early Development, Roche Innovation Center BaselBaselSwitzerland
| | - Peter Bruenker
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | - Christian Klein
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | - Marina Bacac
- Roche Pharma Research & Early Development, Roche Innovation Center ZurichSchlierenSwitzerland
| | - Nikolce Gjorevski
- Roche Pharma Research & Early Development, Roche Innovation Center BaselBaselSwitzerland
| | - Lauriane Cabon
- Roche Pharma Research & Early Development, Roche Innovation Center BaselBaselSwitzerland
| |
Collapse
|
640
|
Kondapalli L, Medina T, Groves DW. Practical cardiovascular imaging approach to diagnose immune checkpoint inhibitor myocarditis. Eur Heart J Cardiovasc Imaging 2021; 22:372-374. [PMID: 33367684 DOI: 10.1093/ehjci/jeaa340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
Immuno-oncology employs various therapeutic strategies that harness a patient's own immune system to fight disease and has been a promising new strategy for cancer therapy over the last decade. Immune checkpoint inhibitors (ICI), are monoclonal antibodies, that increase antitumor immunity by blocking intrinsic down-regulators of immunity, such as cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death 1 (PD-1) or its ligand, programmed cell death ligand 1 (PD-L1). Seven ICIs are currently approved by the Food and Drug Administration and have increased the overall survival for patients with various cancer subtypes. These are used either as single agents or in combination with other checkpoint inhibitors, small molecular kinase inhibitors or cytotoxic chemotherapies. There are also many other immune modifying agents including other checkpoint inhibitor antibodies that are under investigation in clinical trials.
Collapse
Affiliation(s)
- Lavanya Kondapalli
- Division of Cardiology, Department of Medicine, University of Colorado, 12631 E. 17th Avenue, Mail Stop B130, Aurora, CO, USA
| | - Theresa Medina
- Division of Oncology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Daniel W Groves
- Division of Cardiology, Department of Medicine, University of Colorado, 12631 E. 17th Avenue, Mail Stop B130, Aurora, CO, USA.,Division of Cardiothoracic Imaging, Department of Radiology, University of Colorado, Aurora, CO, USA
| |
Collapse
|
641
|
Immunotherapy Treatment for Triple Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:ph14080763. [PMID: 34451860 PMCID: PMC8401402 DOI: 10.3390/ph14080763] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is considered one of the highest-risk subtypes of breast cancer and has dismal prognosis. Local recurrence rate after standard therapy in the early breast cancer setting can be upwards to 72% in 5 years, and in the metastatic setting, the 5-year overall survival is 12%. Due to the lack of receptor expression, there has been a paucity of targeted therapeutics available, with chemotherapy being the primary option for systemic treatment in both the neoadjuvant and metastatic setting. More recently, immunotherapy has revolutionized the landscape of cancer treatment, particularly immune checkpoint inhibitor (ICI) therapy, with FDA approval in over 20 types of cancer since 2011. Compared to other cancer types, breast cancer has been traditionally thought of as being immunologically cold; however, TNBC has demonstrated the most promise with immunotherapy use, a timely discovery due to its lack of targeted therapy options. In this review, we summarize the trials using checkpoint therapy in early and metastatic TNBC, as well as the development of biomarkers and the importance of immune related adverse events (IRAEs), in this disease process.
Collapse
|
642
|
Msaouel P, Oromendia C, Siefker-Radtke AO, Tannir NM, Subudhi SK, Gao J, Wang Y, Siddiqui BA, Shah AY, Aparicio AM, Campbell MT, Zurita AJ, Shaw LK, Lopez LP, McCord H, Chakraborty SN, Perales J, Lu C, Van Alstine ML, Elashoff M, Logothetis C. Evaluation of Technology-Enabled Monitoring of Patient-Reported Outcomes to Detect and Treat Toxic Effects Linked to Immune Checkpoint Inhibitors. JAMA Netw Open 2021; 4:e2122998. [PMID: 34459906 PMCID: PMC8406081 DOI: 10.1001/jamanetworkopen.2021.22998] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IMPORTANCE Immune checkpoint inhibitors can produce distinct toxic effects that require prompt recognition and timely management. OBJECTIVE To develop a technology-enabled, dynamically adaptive protocol that can provide the accurate information needed to inform specific remedies for immune toxic effects in patients treated with immune checkpoint inhibitors. DESIGN, SETTING, AND PARTICIPANTS An open-label cohort study was conducted at a single tertiary referral center from September 6, 2019, to September 3, 2020. The median follow-up duration was 63 (interquartile range, 35.5-122) days. Fifty patients with genitourinary cancers treated with immune checkpoint inhibitors were enrolled. INTERVENTIONS A fit-for-purpose electronic platform was developed to enable active patient and care team participation. A smartphone application downloaded onto patients' personal mobile devices prompted them to report their symptoms at least 3 times per week. The set of symptoms and associated queries were paired with alert thresholds for symptoms requiring clinical action. MAIN OUTCOMES AND MEASURES The primary end point of this interim analysis was feasibility, as measured by patient and care team adherence, and lack of increase in care team staffing. Operating characteristics were estimated for each symptom alert and used to dynamically adapt the alert thresholds to ensure sensitivity while reducing unnecessary alerts. RESULTS Of the 50 patients enrolled, 47 had at least 1 follow-up visit and were included in the analysis. Median age was 65 years (range, 37-86), 39 patients (83%) were men, and 39 patients (83%) had metastatic cancer, with the most common being urothelial cell carcinoma and renal cell carcinoma (22 [47%] patients each). After initial onboarding, no further care team training or additional care team staffing was required. Patients had a median study adherence rate of 74% (interquartile range, 60%-86%) and 73% of automated alerts were reviewed within 3 days by the clinic team. Symptoms with the highest positive predictive value for adverse events requiring acute intervention included dizziness (21%), nausea/vomiting (26%), and shortness of breath (14%). The symptoms most likely to result in unnecessary alerts were arthralgia and myalgia, fatigue, and cough. CONCLUSIONS AND RELEVANCE The findings of this cohort study suggest an acceptable and fiscally sound method can be developed to create a dynamic learning system to detect and manage immune-related toxic effects.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | | | - Arlene O. Siefker-Radtke
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Nizar M. Tannir
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Sumit K. Subudhi
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jianjun Gao
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Yinghong Wang
- Division of Internal Medicine, Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston
| | - Bilal A. Siddiqui
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Amishi Y. Shah
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Ana M. Aparicio
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Matthew T. Campbell
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Amado J. Zurita
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Leah K. Shaw
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Lidia P. Lopez
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Heather McCord
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Sandip N. Chakraborty
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jacqueline Perales
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Cong Lu
- The Ronin Project, San Mateo, California
| | | | | | - Christopher Logothetis
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
643
|
Spyrou N, Vallianou N, Kadillari J, Dalamaga M. The interplay of obesity, gut microbiome and diet in the immune check point inhibitors therapy era. Semin Cancer Biol 2021; 73:356-376. [DOI: 10.1016/j.semcancer.2021.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/22/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
|
644
|
Nelson BE, Hong A, Okereke I, Markowitz A, Willis M, Muthukumarana PV, Nawgiri R. Non-neoplastic inflammatory pseudotumor of the lung after immunotherapy for melanoma: A diagnostic pitfall on fine needle aspiration biopsy of lung. Diagn Cytopathol 2021; 49:1150-1154. [PMID: 34331523 DOI: 10.1002/dc.24846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022]
Abstract
Nivolumab is commonly used as monotherapy or in combination therapy for management of locally advanced or metastatic melanoma; however, it is also associated with immunotherapy-related adverse events concerning for disease progression or tumor flare reaction. This report presents a case of a non-neoplastic pseudotumor of the lung initially mistaken for malignancy that occurred in a patient receiving adjuvant nivolumab therapy following complete resection of stage IIIB melanoma. The diagnosis was made by lung biopsy and confirmed by a wedge resection, with findings consistent with organizing pneumonia type of pulmonary inflammatory pseudotumor rather than malignancy.
Collapse
Affiliation(s)
| | - Angelina Hong
- School of Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Ike Okereke
- Department of Cardiothoracic Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Avi Markowitz
- Department of Hematology & Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Maurice Willis
- Department of Hematology & Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Ranjana Nawgiri
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
645
|
Fuertes MB, Domaica CI, Zwirner NW. Leveraging NKG2D Ligands in Immuno-Oncology. Front Immunol 2021; 12:713158. [PMID: 34394116 PMCID: PMC8358801 DOI: 10.3389/fimmu.2021.713158] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) revolutionized the field of immuno-oncology and opened new avenues towards the development of novel assets to achieve durable immune control of cancer. Yet, the presence of tumor immune evasion mechanisms represents a challenge for the development of efficient treatment options. Therefore, combination therapies are taking the center of the stage in immuno-oncology. Such combination therapies should boost anti-tumor immune responses and/or target tumor immune escape mechanisms, especially those created by major players in the tumor microenvironment (TME) such as tumor-associated macrophages (TAM). Natural killer (NK) cells were recently positioned at the forefront of many immunotherapy strategies, and several new approaches are being designed to fully exploit NK cell antitumor potential. One of the most relevant NK cell-activating receptors is NKG2D, a receptor that recognizes 8 different NKG2D ligands (NKG2DL), including MICA and MICB. MICA and MICB are poorly expressed on normal cells but become upregulated on the surface of damaged, transformed or infected cells as a result of post-transcriptional or post-translational mechanisms and intracellular pathways. Their engagement of NKG2D triggers NK cell effector functions. Also, MICA/B are polymorphic and such polymorphism affects functional responses through regulation of their cell-surface expression, intracellular trafficking, shedding of soluble immunosuppressive isoforms, or the affinity of NKG2D interaction. Although immunotherapeutic approaches that target the NKG2D-NKG2DL axis are under investigation, several tumor immune escape mechanisms account for reduced cell surface expression of NKG2DL and contribute to tumor immune escape. Also, NKG2DL polymorphism determines functional NKG2D-dependent responses, thus representing an additional challenge for leveraging NKG2DL in immuno-oncology. In this review, we discuss strategies to boost MICA/B expression and/or inhibit their shedding and propose that combination strategies that target MICA/B with antibodies and strategies aimed at promoting their upregulation on tumor cells or at reprograming TAM into pro-inflammatory macrophages and remodeling of the TME, emerge as frontrunners in immuno-oncology because they may unleash the antitumor effector functions of NK cells and cytotoxic CD8 T cells (CTL). Pursuing several of these pipelines might lead to innovative modalities of immunotherapy for the treatment of a wide range of cancer patients.
Collapse
Affiliation(s)
- Mercedes Beatriz Fuertes
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Carolina Inés Domaica
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Norberto Walter Zwirner
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
646
|
Waldron JL, Schworer SA, Kwan M. Hypersensitivity and Immune-related Adverse Events in Biologic Therapy. Clin Rev Allergy Immunol 2021; 62:413-431. [PMID: 34319562 DOI: 10.1007/s12016-021-08879-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 12/13/2022]
Abstract
Biologic medications are an expanding field of therapeutics for various medical conditions including cancer and inflammatory diseases. Due to their targeted approach to therapy, biologics can be less toxic than traditional systemic medications. However, as use becomes more widespread, adverse effects from biologic administration have also become apparent. Immune-related adverse events are a common mechanism by which biologics can cause on-target immune-related toxicities and both immediate and delayed-type hypersensitivity reactions. Immediate hypersensitivity reactions can be mediated by cytokine release or antibody mediated reactions, while delayed-type hypersensitivity is most often caused by serum sickness-like reactions. Additionally, biologics used for treatment of cancer using checkpoint blockade and rheumatologic disease using cytokine blockade can result in autoimmunity. Finally, when inflammatory cytokines are targeted for treatment of autoimmune or autoinflammatory disease, the host immune defense can be compromised predisposing to secondary immunodeficiency. This review will discuss the mechanisms of these reactions and discuss examples of biologics implicated in each of these adverse events.
Collapse
Affiliation(s)
- Jamie L Waldron
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Stephen A Schworer
- Department of Pediatrics, Division of Allergy & Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Mildred Kwan
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, UNC School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
647
|
Sullivan RJ, Weber JS. Immune-related toxicities of checkpoint inhibitors: mechanisms and mitigation strategies. Nat Rev Drug Discov 2021; 21:495-508. [PMID: 34316029 DOI: 10.1038/s41573-021-00259-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2021] [Indexed: 02/07/2023]
Abstract
The immune-related adverse events associated with treatment with immune checkpoint inhibitors result in significant morbidity for patients as well as considerable cost to the health-care system, and can limit the use of these beneficial drugs. Understanding the mechanisms of these side effects and how they can be separated from the antitumour effects of immune checkpoint inhibitors, as well as identifying biomarkers that predict the development of immune-related toxicities, will facilitate the conduct of trials to limit their onset and improve patient outcomes. In this Review, we discuss the different types of immune-related adverse events and how their treatment and identification of possible predictive biomarkers may shed light on their mechanisms, and describe possible strategies and targets for prophylactic and therapeutic intervention to mitigate them.
Collapse
Affiliation(s)
- Ryan J Sullivan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Comprehensive Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
648
|
He S, Li J, Cheng P, Zeng Z, Zhang C, Duan H, Pu K. Charge-Reversal Polymer Nano-modulators for Photodynamic Immunotherapy of Cancer. Angew Chem Int Ed Engl 2021; 60:19355-19363. [PMID: 34105217 DOI: 10.1002/anie.202106392] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Indexed: 12/19/2022]
Abstract
Nanomedicine can regulate the balance between cytotoxic T lymphocytes (CTLs) and suppressive regulatory T lymphocytes (Tregs), which however has been rarely exploited for cancer immunotherapy. We report a charge-reversal polymer nano-modulator (SPDMC N) activated by tumor microenvironment (TME) for photodynamic immunotherapy of cancer. SPDMC N is constructed by conjugating an immunomodulator (demethylcantharidin, DMC) to the side chains of a photodynamic polymer via an acid-liable linker. The negative charge of SPDMC N ensures its high stability in blood circulation and ideal tumor accumulation; exposure to acidic TME reverses its surface charge to positive, enhancing tumor penetration and locally releasing DMC. Upon near-infrared photoirradiation, SPDMC N generates singlet oxygen to ablate tumors and promote maturation of dendritic cells. Released DMC inhibits protein phosphatase 2 (PP2A) activity and decreases Tregs differentiation. Such combinational action induces a sharp increase in CTL/Treg ratio in TME and effectively inhibits both primary and distant tumors in living mice.
Collapse
Affiliation(s)
- Shasha He
- School of Chemical and Biomedical Engineering, School of Physical and Mathematical Sciences, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jingchao Li
- School of Chemical and Biomedical Engineering, School of Physical and Mathematical Sciences, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Penghui Cheng
- School of Chemical and Biomedical Engineering, School of Physical and Mathematical Sciences, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Ziling Zeng
- School of Chemical and Biomedical Engineering, School of Physical and Mathematical Sciences, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Chi Zhang
- School of Chemical and Biomedical Engineering, School of Physical and Mathematical Sciences, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering, School of Physical and Mathematical Sciences, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, School of Physical and Mathematical Sciences, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| |
Collapse
|
649
|
Tang XY, Shi AP, Xiong YL, Zheng KF, Liu YJ, Shi XG, Jiang T, Zhao JB. Clinical Research on the Mechanisms Underlying Immune Checkpoints and Tumor Metastasis. Front Oncol 2021; 11:693321. [PMID: 34367975 PMCID: PMC8339928 DOI: 10.3389/fonc.2021.693321] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/07/2021] [Indexed: 12/13/2022] Open
Abstract
This study highlights aspects of the latest clinical research conducted on the relationship between immune checkpoints and tumor metastasis. The overview of each immune checkpoint is divided into the following three sections: 1) structure and expression; 2) immune mechanism related to tumor metastasis; and 3) clinical research related to tumor metastasis. This review expands on the immunological mechanisms of 17 immune checkpoints, including TIM-3, CD47, and OX-40L, that mediate tumor metastasis; evidence shows that most of these immune checkpoints are expressed on the surface of T cells, which mainly exert immunomodulatory effects. Additionally, we have summarized the roles of these immune checkpoints in the diagnosis and treatment of metastatic tumors, as these checkpoints are considered common predictors of metastasis in various cancers such as prostate cancer, non-Hodgkin lymphoma, and melanoma. Moreover, certain immune checkpoints can be used in synergy with PD-1 and CTLA-4, along with the implementation of combination therapies such as LIGHT-VTR and anti-PD-1 antibodies. Presently, most monoclonal antibodies generated against immune checkpoints are under investigation as part of ongoing preclinical or clinical trials conducted to evaluate their efficacy and safety to establish a better combination treatment strategy; however, no significant progress has been made regarding monoclonal antibody targeting of CD28, VISTA, or VTCN1. The application of immune checkpoint inhibitors in early stage tumors to prevent tumor metastasis warrants further evidence; the immune-related adverse events should be considered before combination therapy. This review aims to elucidate the mechanisms of immune checkpoint and the clinical progress on their use in metastatic tumors reported over the last 5 years, which may provide insights into the development of novel therapeutic strategies that will assist with the utilization of various immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Xi-Yang Tang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - An-Ping Shi
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, China
| | - Yan-Lu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Kai-Fu Zheng
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Yu-Jian Liu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Xian-Gui Shi
- College of Basic Medicine, Air Force Medical University, Xi’an, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Jin-Bo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
650
|
Ishiyama Y, Kondo T, Nemoto Y, Kobari Y, Ishihara H, Tachibana H, Yoshida K, Hashimoto Y, Takagi T, Iizuka J, Tanabe K. Antibiotic use and survival of patients receiving pembrolizumab for chemotherapy-resistant metastatic urothelial carcinoma. Urol Oncol 2021; 39:834.e21-834.e28. [PMID: 34284929 DOI: 10.1016/j.urolonc.2021.05.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The use of antibiotics alters gut microbiota and has been reported to impact outcomes in immune checkpoint inhibitor (ICI) treatment in various types of cancer. We investigated the impact of antibiotics on patients with metastatic urothelial carcinoma (mUC) treated with pembrolizumab. MATERIALS AND METHODS The data of 67 patients with chemotherapy-resistant mUC who were treated with pembrolizumab were retrospectively evaluated. The patients were classified into groups according to antibiotic status (with-antibiotic and without-antibiotic), and the progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and disease control rate (DCR) were compared between the 2 groups. RESULTS PFS (median: 1.1 vs. 8.9 months; P < 0.001) and OS (median: 2.3 vs. 19.5 months; P < 0.001) were significantly shorter in the with-antibiotic group (n = 15, 22%) than in the without-antibiotic group (n = 52, 78%). Patients in the with-antibiotic group had significantly higher Eastern Cooperative Oncology Group performance status scores (P = 0.042). Multivariable analyses revealed antibiotic use as an independent predictor of PFS (P < 0.001) and OS (P = 0.002). No patients in the with-antibiotic group achieved a complete response to pembrolizumab. The ORR (complete response (CR) + partial response (PR)) was higher among patients not treated with antibiotics than among patients treated with antibiotics, though the difference was not significant (34.6% vs. 13.3%, P = 0.093). The DCR (CR + PR + stable disease) was also higher among patients in the with-antibiotic group than in the without-antibiotic group (57.7% vs. 20.0%, P = 0.008). CONCLUSION The use of antibiotics was negatively associated with outcomes in patients with mUC who are administered pembrolizumab. Baseline performance status was worse for these patients. Further analyses are required to identify associations between antibiotic use, bacterial infection for which it was indicated or its influence on performance status, on treatment outcomes.
Collapse
Affiliation(s)
- Yudai Ishiyama
- Department of Urology, Tokyo Women's Medical University Medical Center East, Arakawa-ku, Tokyo, Japan; Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Tsunenori Kondo
- Department of Urology, Tokyo Women's Medical University Medical Center East, Arakawa-ku, Tokyo, Japan.
| | - Yuki Nemoto
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan; Department of Urology, Saiseikai Kawaguchi General Hospital, Kawaguchi, Saitama, Japan
| | - Yuki Kobari
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Hiroki Ishihara
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Hidekazu Tachibana
- Department of Urology, Tokyo Women's Medical University Medical Center East, Arakawa-ku, Tokyo, Japan
| | - Kazuhiko Yoshida
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yasunobu Hashimoto
- Department of Urology, Saiseikai Kawaguchi General Hospital, Kawaguchi, Saitama, Japan
| | - Toshio Takagi
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Junpei Iizuka
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Kazunari Tanabe
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|