Opportunistic Infections and Immune-Related Adverse Events Associated with Administering Immune Checkpoint Inhibitors: A Narrative Review
Abstract
:1. Introduction
- ICIs increase T cell activation against antigens expressed in tumour cells and healthy tissue.
- ICIs increase levels of pre-existent auto-antibodies.
- ICIs increase the level of inflammatory cytokines.
- Anti-CTLA-4 antibodies cause complement-mediated inflammation, impairing normal tissue that expresses CTLA-4.
2. Immune Checkpoints: How Do They Induce Inhibition of Activated T Cells?
3. IMT and Cancer: Successful Relation
Drug | Target | Type | Refs. |
---|---|---|---|
Nivolumab | PD-1 | Human IgG4, anti-PD-1 | [39,40,41] |
Pembrolizumab | PD-1 | Humanized IgG4, anti-PD-1 | [37,42,43,44] |
Atezolizumab | PD-L1 | Humanized IgG1, anti-PD-L1 | [45,46,47] |
Cemiplimab | PD-1 | Human IgG4, anti-PD-1 | [48,49] |
Ipilimumab | CTLA-4 | Human IgG1, anti-CTLA-4 | [8,50,51] |
Avelumab | PD-L1 | Human IgG1 anti-PD-L1 | [52,53,54,55] |
Durvalumab | PD-L1 | Human IgG1κ anti-PD-L1 | [7,56,57] |
Dostarlimab | PD-1 | Humanized IgG4 anti-PD-1 | [58,59,60] |
Tremelimumab | CTLA-4 | Human IgG2 anti-CTLA-4 | [56] |
Relatlimab | LAG-3 | Human IgG4 anti-LAG-3 | [9,61] |
4. Infectious Diseases Associated with the Use of IMT
4.1. Tuberculosis (TB)
4.2. Aspergillus fumigatus Infection
4.3. Pneumocystis jirovecii
5. Organ Damage by the Use of IMT, Even in the Absence of Pathogens
5.1. Adverse Hepatic Events
5.2. Pulmonary Adverse Events
5.3. Dermatological Toxicity Events
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Twomey, J.D.; Zhang, B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J. 2021, 23, 39. [Google Scholar] [CrossRef] [PubMed]
- Egen, J.G.; Kuhns, M.S.; Allison, J.P. CTLA-4: New insights into its biological function and use in tumor immunotherapy. Nat. Immunol. 2002, 3, 611–618. [Google Scholar] [CrossRef] [PubMed]
- Yu, W.; Hua, Y.; Qiu, H.; Hao, J.; Zou, K.; Li, Z.; Hu, S.; Guo, P.; Chen, M.; Sui, S.; et al. PD-L1 promotes tumor growth and progression by activating WIP and β-catenin signaling pathways and predicts poor prognosis in lung cancer. Cell Death Dis. 2020, 11, 506. [Google Scholar] [CrossRef]
- Li, F.; Li, N.; Sang, J.; Fan, X.; Deng, H.; Zhang, X.; Han, Q.; Lv, Y.; Liu, Z. Highly elevated soluble Tim-3 levels correlate with increased hepatocellular carcinoma risk and poor survival of hepatocellular carcinoma patients in chronic hepatitis B virus infection. Cancer Manag. Res. 2018, 10, 941–951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Contardi, E.; Palmisano, G.L.; Tazzari, P.L.; Martelli, A.M.; Falà, F.; Fabbi, M.; Kato, T.; Lucarelli, E.; Donati, D.; Polito, L.; et al. CTLA-4 is constitutively expressed on tumor cells and can trigger apoptosis upon ligand interaction. Int. J. Cancer 2005, 117, 538–550. [Google Scholar] [CrossRef] [PubMed]
- Kato, T.; Masuda, N.; Nakanishi, Y.; Takahashi, M.; Hida, T.; Sakai, H.; Atagi, S.; Fujita, S.; Tanaka, H.; Takeda, K.; et al. Nivolumab-induced interstitial lung disease analysis of two phase II studies patients with recurrent or advanced non-small-cell lung cancer. Lung Cancer 2016, 104, 111–118. [Google Scholar] [CrossRef] [Green Version]
- Paz-Ares, L.; Spira, A.; Raben, D.; Planchard, D.; Cho, B.C.; Özgüroğlu, M.; Daniel, D.; Villegas, A.; Vicente, D.; Hui, R.; et al. Outcomes with durvalumab by tumour PD-L1 ex-pression in unresectable, stage III non-small-cell lung cancer in the PACIFIC trial. Ann. Oncol. 2020, 31, 798–806. [Google Scholar] [CrossRef]
- Wolchok, J.D.; Neyns, B.; Linette, G.; Negrier, S.; Lutzky, J.; Thomas, L.; Waterfield, W.; Schadendorf, D.; Smylie, M.; Guthrie, T., Jr.; et al. Ipilimumab monotherapy in patients with pretreated advanced melanoma: A randomized, double-blind, multicentre, phase 2, dose-ranging study. Lancet Oncol. 2010, 11, 155–164. [Google Scholar] [CrossRef]
- Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Gutiérrez, E.C.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef]
- Huang, Y.-H.; Zhu, C.; Kondo, Y.; Anderson, A.C.; Gandhi, A.; Russell, A.F.; Dougan, S.K.; Petersen, B.-S.; Melum, E.; Pertel, T.; et al. CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature 2015, 517, 386–390. [Google Scholar] [CrossRef]
- Wherry, E.J.; Ha, S.-J.; Kaech, S.M.; Haining, W.N.; Sarkar, S.; Kalia, V.; Subramaniam, S.; Blattman, J.N.; Barber, D.L.; Ahmed, R. Molecular Signature of CD8+ T Cell Exhaustion during Chronic Viral Infection. Immunity 2007, 27, 670–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beldi-Ferchiou, A.; Lambert, M.; Dogniaux, S.; Vély, F.; Vivier, E.; Olive, D.; Dupuy, S.; Levasseur, F.; Zucman, D.; Lebbé, C.; et al. PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget 2016, 7, 72961–72977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rallón, N.; García, M.; García-Samaniego, J.; Cabello, A.; Álvarez, B.; Restrepo, C.; Nistal, S.; Górgolas, M.; Benito, J.M. Expression of PD-1 and Tim-3 markers of T-cell exhaustion is associated with CD4 dynamics during the course of untreated and treated HIV infection. PLoS ONE 2018, 13, e0193829. [Google Scholar] [CrossRef] [PubMed]
- Golden-Mason, L.; Palmer, B.E.; Kassam, N.; Townshend-Bulson, L.; Livingston, S.; McMahon, B.J.; Castelblanco, N.; Kuchroo, V.; Gretch, D.R.; Rosen, H.R. Faculty Opinions recommendation of Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J. Virol. 2009, 83, 9122–9130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, R.B.; Ndhlovu, L.C.; Barbour, J.D.; Sheth, P.M.; Jha, A.R.; Long, B.R.; Wong, J.C.; Satkunarajah, M.; Schweneker, M.; Chapman, J.M.; et al. Tim-3 expression defines a novel population of dys-functional T cells with highly elevated frequencies in progressive HIV-1 infection. J. Exp. Med. 2008, 205, 2763–2779. [Google Scholar] [CrossRef] [PubMed]
- Tivol, E.A.; Borriello, F.; Schweitzer, A.N.; Lynch, W.P.; Bluestone, J.A.; Sharpe, A.H. Loss of CTLA-4 leads to massive lymphoprolif-eration and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 1995, 3, 541–547. [Google Scholar] [CrossRef] [Green Version]
- Nishimura, H.; Nose, M.; Hiai, H.; Minato, N.; Honjo, T. Development of Lupus-like Autoimmune Diseases by Disruption of the PD-1 Gene Encoding an ITIM Motif-Carrying Immunoreceptor. Immunity 1999, 11, 141–151. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Okazaki, I.-M.; Yoshida, T.; Chikuma, S.; Kato, Y.; Nakaki, F.; Hiai, H.; Honjo, T.; Okazaki, T. PD-1 deficiency results in the development of fatal myocarditis in MRL mice. Int. Immunol. 2010, 22, 443–452. [Google Scholar] [CrossRef]
- Postow, M.A.; Sidlow, R.; Hellmann, M.D. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N. Engl. J. Med. 2018, 378, 158–168. [Google Scholar] [CrossRef]
- Wucherpfennig, K.W.; Gagnon, E.; Huseby, E.S.; Call, M.E. Structural Biology of the T-cell Receptor: Insights into Receptor Assembly, Ligand Recognition, and Initiation of Signaling. Cold Spring Harb. Perspect. Biol. 2010, 2, a005140. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, T.D.; Carrascal, M.; Vidal-Cortes, O.; Gallardo, O.; Casas, V.; Gay, M.; Phan, V.C.; Abian, J. The phosphoproteome of human Jurkat T cell clones upon co-stimulation with anti-CD3/anti-CD28 antibodies. J. Proteomics 2016, 131, 190–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thiel, M.; Wolfs, M.; Bauer, S.; Wenning, A.S.; Burckhart, T.; Schwarz, E.C.; Scott, A.M.; Renner, C.; Hoth, M. Efficiency of T-cell costimulation by CD80 and CD86 cross-linking correlates with calcium entry. Immunology 2009, 129, 28–40. [Google Scholar] [CrossRef] [PubMed]
- Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene super-family, upon programmed cell death. EMBO J. 1992, 11, 3887–3895. [Google Scholar] [CrossRef]
- del Rio, M.-L.; Seebach, J.D.; Fernández-Renedo, C.; Rodriguez-Barbosa, J.-I. ITIM-dependent negative signaling pathways for the control of cell-mediated xenogeneic immune responses. Xenotransplantation 2013, 20, 397–406. [Google Scholar] [CrossRef] [PubMed]
- Van Der Merwe, P.A.; Bodian, D.L.; Daenke, S.; Linsley, P.; Davis, S.J. CD80 (B7-1) Binds Both CD28 and CTLA-4 with a Low Affinity and Very Fast Kinetics. J. Exp. Med. 1997, 185, 393–404. [Google Scholar] [CrossRef]
- Alegre, M.-L.; Frauwirth, K.A.; Thompson, C.B. T-cell regulation by CD28 and CTLA-4. Nat. Rev. Immunol. 2001, 1, 220–228. [Google Scholar] [CrossRef]
- Triebel, F.; Jitsukawa, S.; Baixeras, E.; Roman-Roman, S.; Genevee, C.; Viegas-Pequignot, E.; Hercend, T. LAG-3, a novel lymphocyte acti-vation gene closely related to CD4. J. Exp. Med. 1990, 171, 1393–1405. [Google Scholar] [CrossRef]
- Workman, C.J.; Rice, D.S.; Dugger, K.J.; Kurschner, C.; Vignali, D.A.A. Phenotypic analysis of the murine CD4-related glycoprotein, CD223 (LAG-3). Eur. J. Immunol. 2002, 32, 2255–2263. [Google Scholar] [CrossRef]
- Shang, Y.; Li, Z.; Li, H.; Xia, H.; Lin, Z. TIM-3 expression in human osteosarcoma: Correlation with the expression of epitheli-al-mesenchymal transition-specific biomarkers. Oncol. Lett. 2013, 6, 490–494. [Google Scholar] [CrossRef] [Green Version]
- Cao, Y.; Zhou, X.; Huang, X.; Li, Q.; Gao, L.; Jiang, L.; Huang, M.; Zhou, J. Tim-3 Expression in Cervical Cancer Promotes Tumor Metastasis. PLoS ONE 2013, 8, e53834. [Google Scholar] [CrossRef]
- Ma, C.J.; Li, G.Y.; Cheng, Y.Q.; Wang, J.M.; Ying, R.S.; Shi, L.; Wu, X.Y.; Niki, T.; Hirashima, M.; Li, C.F.; et al. Cis Association of Galectin-9 with Tim-3 Differentially Regulates IL-12/IL-23 Expressions in Monocytes via TLR Signaling. PLoS ONE 2013, 8, e72488. [Google Scholar] [CrossRef] [Green Version]
- Rangachari, M.; Zhu, C.; Sakuishi, K.; Xiao, S.; Karman, J.; Chen, A.; Angin, M.; Wakeham, A.; Greenfield, E.A.; Sobel, R.A.; et al. Bat3 promotes T cell responses and autoimmunity by repressing Tim-3–mediated cell death and exhaustion. Nat. Med. 2012, 18, 1394–1400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.; Su, E.W.; Zhu, C.; Hainline, S.; Phuah, J.; Moroco, J.A.; Smithgall, T.E.; Kuchroo, V.K.; Kane, L.P. Phosphotyrosine-Dependent Coupling of Tim-3 to T-Cell Receptor Signaling Pathways. Mol. Cell. Biol. 2011, 31, 3963–3974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hargadon, K.M.; Johnson, C.E.; Williams, C.J. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 2018, 62, 29–39. [Google Scholar] [CrossRef]
- Hu, H.; Wang, K.; Jia, R.; Zeng, Z.-X.; Zhu, M.; Deng, Y.-L.; Xiong, Z.-J.; Tang, J.-N.; Xie, H.; Wang, Y.; et al. Current Status in Rechallenge of Immunotherapy. Int. J. Biol. Sci. 2023, 19, 2428–2442. [Google Scholar] [CrossRef] [PubMed]
- Cercek, A.; Lumish, M.; Sinopoli, J.; Weiss, J.; Shia, J.; Lamendola-Essel, M.; El Dika, I.H.; Segal, N.; Shcherba, M.; Sugarman, R.; et al. PD-1 Blockade in Mismatch Repair–Deficient, Locally Advanced Rectal Cancer. N. Engl. J. Med. 2022, 386, 2363–2376. [Google Scholar] [CrossRef] [PubMed]
- André, T.; Shiu, K.-K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Garcia-Carbonero, R.; Benavides, M.; Gibbs, P.; et al. Pembrolizumab in Microsatellite-Instability–High Advanced Colorectal Cancer. N. Engl. J. Med. 2020, 383, 2207–2218. [Google Scholar] [CrossRef] [PubMed]
- Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.-P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients with Noncolorectal High Microsatellite Instability/Mismatch Repair–Deficient Cancer: Results from the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef]
- Brahmer, J.R.; Hammers, H.; Lipson, E.J. Nivolumab: Targeting PD-1 to bolster antitumor immunity. Futur. Oncol. 2015, 11, 1307–1326. [Google Scholar] [CrossRef]
- Ansell, S.M.; Lesokhin, A.M.; Borrello, I.; Halwani, A.; Scott, E.C.; Gutierrez, M.; Schuster, S.J.; Millenson, M.M.; Cattry, D.; Freeman, G.J.; et al. PD-1 Blockade with Nivolumab in Relapsed or Refractory Hodgkin’s Lymphoma. N. Engl. J. Med. 2015, 372, 311–319. [Google Scholar] [CrossRef] [Green Version]
- Hodi, F.S.; Postow, M.A.; Chesney, J.A.; Pavlick, A.C.; Robert, C.; Grossmann, K.F.; McDermott, D.F.; Linette, G.P.; Meyer, N.; Giguere, J.K.; et al. Clinical response, progression-free survival (PFS), and safety in patients (pts) with advanced melanoma (MEL) receiving nivolumab (NIVO) combined with ipilimumab (IPI) vs IPI monotherapy in CheckMate 069 study. J. Clin. Oncol. 2015, 33, 9004. [Google Scholar] [CrossRef]
- Wu, G. Therapeutic Effects of Pembrolizumab Combined with Paclitaxel and Cisplatin Chemotherapy on Advanced Non-Squamous Non-Small Cell Lung Cancer and Influencing Factors. Indian J. Pharm. Sci. 2021, 83, 120–126. [Google Scholar] [CrossRef]
- Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the Treatment of Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef] [PubMed]
- Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [Green Version]
- Li, J.-X.; Huang, J.-M.; Jiang, Z.-B.; Li, R.-Z.; Sun, A.; Leung, E.L.-H.; Yan, P.-Y. Current Clinical Progress of PD-1/PD-L1 Immunotherapy and Potential Combination Treatment in Non–Small Cell Lung Cancer. Integr. Cancer Ther. 2019, 18, 1534735419890020. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Yang, D.; Min, Y.; Liao, A.; Zhao, J.; Jiang, L.; Dong, X.; Deng, W.; Yu, H.; Yu, R.; et al. First-line atezolizumab/durvalumab plus platinum–etoposide combined with radiotherapy in extensive-stage small-cell lung cancer. BMC Cancer 2023, 23, 318. [Google Scholar] [CrossRef]
- Wang, K.; Liu, Z.-H.; Yu, H.-M.; Cheng, Y.-Q.; Xiang, Y.-J.; Zhong, J.-Y.; Ni, Q.-Z.; Zhou, L.-P.; Liang, C.; Zhou, H.-K.; et al. Efficacy and safety of a triple combination of atezolizumab, bevacizumab plus GEMOX for advanced biliary tract cancer: A multicenter, single-arm, retrospective study. Ther. Adv. Gastroenterol. 2023, 16, 17562848231160630. [Google Scholar] [CrossRef]
- Migden, M.R.; Rischin, D.; Schmults, C.D.; Guminski, A.; Hauschild, A.; Lewis, K.D.; Chung, C.H.; Hernandez-Aya, L.F.; Lim, A.M.; Chang, A.L.S.; et al. PD-1 Blockade with Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. N. Engl. J. Med. 2018, 379, 341–351. [Google Scholar] [CrossRef] [Green Version]
- Lesokhin, A.; LeBlanc, R.; Dimopoulos, M.A.; Capra, M.; Carlo-Stella, C.; Karlin, L.; Castilloux, J.; Forsberg, P.; Parmar, G.; Tosikyan, A.; et al. Isatuximab in combination with cemiplimab in patients with relapsed/refractory multiple myeloma: A phase 1/2 study. Cancer Med. 2023, 12, 10254–10266. [Google Scholar] [CrossRef]
- Mansh, M. Ipilimumab and cancer immunotherapy: A new hope for advanced stage melanoma. Yale J. Biol. Med. 2011, 84, 381–389. [Google Scholar]
- O’Day, S.J.; Maio, M.; Chiarion-Sileni, V.; Gajewski, T.F.; Pehamberger, H.; Bondarenko, I.N.; Queirolo, P.; Lundgren, L.; Mikhailov, S.; Roman, L.; et al. Efficacy and safety of ipilimumab monotherapy in patients with pretreated advanced melanoma: A multicenter single-arm phase II study. Ann. Oncol. 2010, 21, 1712–1717. [Google Scholar] [CrossRef] [PubMed]
- Shirley, M. Avelumab: A Review in Metastatic Merkel Cell Carcinoma. Target. Oncol. 2018, 13, 409–416. [Google Scholar] [CrossRef] [PubMed]
- Averbuch, I.; Stoff, R.; Miodovnik, M.; Fennig, S.; Bar-Sela, G.; Yakobson, A.; Daliot, J.; Asher, N.; Fenig, E. Avelumab for the treatment of locally advanced or metastatic Merkel cell carcinoma—A multicenter real-world experience in Israel. Cancer Med. 2023, 12, 12065–12070. [Google Scholar] [CrossRef] [PubMed]
- Eyck, J.E.T.; Kahlon, N.; Masih, S.; Hamouda, D.M.; Petros, F.G. Clinical Evaluation of Avelumab in the Treatment of Advanced Urothelial Carcinoma: Focus on Patient Selection and Outcomes. Cancer Manag. Res. 2022, 14, 729–738. [Google Scholar] [CrossRef]
- Li, H.; Sahu, K.K.; Brundage, J.; Benson, M.; Swami, U.; Boucher, K.M.; Gupta, S.; Hawks, J.; Sirohi, D.; Agarwal, N.; et al. Phase I Trial of Combination Therapy with Avelumab and Cabozantinib in Patients With Newly Diagnosed Metastatic Clear Cell Renal Cell Carcinoma. Oncologist 2023, 28, 737-e693. [Google Scholar] [CrossRef]
- Abou-Alfa, G.K.; Lau, G.; Kudo, M.; Chan, S.L.; Kelley, R.K.; Furuse, J.; Sukeepaisarnjaroen, W.; Kang, Y.-K.; Van Dao, T.; De Toni, E.N.; et al. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma. NEJM Evid. 2022, 1–12. [Google Scholar] [CrossRef]
- Cortiula, F.; De Ruysscher, D.; Steens, M.; Wijsman, R.; van der Wekken, A.; Alberti, M.; Hendriks, L.E. Adjuvant durvalumab after concurrent chemoradiotherapy for patients with unresectable stage III NSCLC harbouring uncommon genomic alterations. Eur. J. Cancer 2023, 184, 172–178. [Google Scholar] [CrossRef]
- Kuchimanchi, M.; Dabrowski, C.; Lu, S.; Melhem, M. Dostarlimab, an anti-programmed death-1 monoclonal antibody, does not cause QT prolongation in patients with solid tumours: A concentration-QT analysis. Br. J. Clin. Pharmacol. 2023, 89, 2272–2282. [Google Scholar] [CrossRef]
- Madariaga, A.; Garg, S.; Tchrakian, N.; Dhani, N.C.; Jimenez, W.; Welch, S.; MacKay, H.; Ethier, J.-L.; Gilbert, L.; Li, X.; et al. Clinical outcome and biomarker assessments of a multi-centre phase II trial assessing niraparib with or without dostarlimab in recurrent endometrial carcinoma. Nat. Commun. 2023, 14, 1452. [Google Scholar] [CrossRef]
- Oaknin, A.; Gilbert, L.; Tinker, A.V.; Brown, J.; Mathews, C.; Press, J.; Sabatier, R.; O’malley, D.M.; Samouelian, V.; Boni, V.; et al. Safety and antitumor activity of dostarlimab in patients with advanced or recurrent DNA mismatch repair deficient/microsatellite instability-high (dMMR/MSI-H) or proficient/stable (MMRp/MSS) endometrial cancer: Interim results from GARNET—A phase I, single-arm study. J. Immunother. Cancer 2022, 10, e003777. [Google Scholar] [CrossRef]
- Sordo-bahamonde, C.; Lorenzo-herrero, S.; González-rodríguez, A.P.; Payer, Á.R.; González-garcía, E.; López-soto, A.; Gonzalez, S. Lag-3 blockade with relatlimab (Bms-986016) restores anti-leukemic responses in chronic lymphocytic leukemia. Cancers 2021, 13, 2112. [Google Scholar] [CrossRef] [PubMed]
- Morelli, T.; Fujita, K.; Redelman-Sidi, G.; Elkington, P.T. Infections due to dysregulated immunity: An emerging complication of cancer immunotherapy. Thorax 2021, 77, 304–311. [Google Scholar] [CrossRef]
- Inthasot, V.; Bruyneel, M.; Muylle, I.; Ninane, V. Severe pulmonary infections complicating nivolumab treatment for lung cancer: A report of two cases. Acta Clin. Belg. 2019, 75, 308–310. [Google Scholar] [CrossRef] [PubMed]
- Beham, A.W.; Puellmann, K.; Laird, R.; Fuchs, T.; Streich, R.; Breysach, C.; Raddatz, D.; Oniga, S.; Peccerella, T.; Findeisen, P.; et al. A TNF-Regulated Recombinatorial Macrophage Immune Receptor Implicated in Granuloma Formation in Tuberculosis. PLOS Pathog. 2011, 7, e1002375. [Google Scholar] [CrossRef] [Green Version]
- Robert, M.; Miossec, P. Reactivation of latent tuberculosis with TNF inhibitors: Critical role of the beta 2 chain of the IL-12 receptor. Cell. Mol. Immunol. 2021, 18, 1644–1651. [Google Scholar] [CrossRef] [PubMed]
- Palacios, Y.; Chavez-Galan, L. Immunosuppressant Therapies in COVID-19: Is the TNF Axis an Alternative? Pharmaceuticals 2022, 15, 616. [Google Scholar] [CrossRef]
- Keane, J.; Gershon, S.; Wise, R.P.; Mirabile-Levens, E.; Kasznica, J.; Schwieterman, W.D.; Siegel, J.N.; Braun, M.M. Tuberculosis Associated with Imfliximab, a Tumor Necrosis Factor Alpha-Neutralizing Agent. Infect. Dis. Clin. Pract. 2001, 10, 498. [Google Scholar] [CrossRef]
- Lázár-Molnár, E.; Chen, B.; Sweeney, K.A.; Wang, E.J.; Liu, W.; Lin, J.; Porcelli, S.A.; Almo, S.C.; Nathenson, S.G.; Jacobs, W.R. Programmed death-1 (PD-1)–deficient mice are extraordinarily sensitive to tuberculosis. Proc. Natl. Acad. Sci. USA 2010, 107, 13402–13407. [Google Scholar] [CrossRef]
- Barber, D.L.; Sakai, S.; Kudchadkar, R.R.; Fling, S.P.; Day, T.A.; Vergara, J.A.; Ashkin, D.; Cheng, J.H.; Lundgren, L.M.; Raabe, V.N.; et al. Tuberculosis following PD-1 blockade for cancer immunotherapy. Sci. Transl. Med. 2019, 11, eaat2702. [Google Scholar] [CrossRef]
- Barber, D.L.; Mayer-Barber, K.D.; Feng, C.G.; Sharpe, A.H.; Sher, A. CD4 T Cells Promote Rather than Control Tuberculosis in the Absence of PD-1–Mediated Inhibition. J. Immunol. 2011, 186, 1598–1607. [Google Scholar] [CrossRef] [Green Version]
- Suliman, A.M.; Bek, S.A.; Elkhatim, M.S.; Husain, A.A.; Mismar, A.Y.; Eldean, M.Z.S.; Lengyel, Z.; Elazzazy, S.; Rasul, K.I.; Omar, N.E. Tuberculosis following programmed cell death receptor-1 (PD-1) inhibitor in a patient with non-small cell lung cancer. Case report and literature review. Cancer Immunol. Immunother. 2020, 70, 935–944. [Google Scholar] [CrossRef] [PubMed]
- Bae, S.; Kim, Y.-J.; Kim, M.-J.; Kim, J.H.; Yun, S.-C.; Jung, J.; Chong, Y.P.; Kim, S.-H.; Choi, S.-H.; Kim, Y.S.; et al. Risk of tuberculosis in patients with cancer treated with immune checkpoint inhibitors: A nationwide observational study. J. Immunother. Cancer 2021, 9, e002960. [Google Scholar] [CrossRef] [PubMed]
- Liu, K.; Wang, D.; Yao, C.; Qiao, M.; Li, Q.; Ren, W.; Li, S.; Gao, M.; Pang, Y. Increased Tuberculosis Incidence Due to Immunotherapy Based on PD-1 and PD-L1 Blockade: A Systematic Review and Meta-Analysis. Front. Immunol. 2022, 13, 727220. [Google Scholar] [CrossRef] [PubMed]
- O’gorman, C.M. Airborne Aspergillus fumigatus conidia: A risk factor for aspergillosis. Fungal Biol. Rev. 2011, 25, 151–157. [Google Scholar] [CrossRef]
- Knutsen, A.P.; Slavin, R.G. Allergic Bronchopulmonary Aspergillosis in Asthma and Cystic Fibrosis. J. Immunol. Res. 2011, 2011, 843763. [Google Scholar] [CrossRef] [Green Version]
- Malek, A.E.; Taremi, M.; Spallone, A.; Alvarez-Cardona, J.J.; Kontoyiannis, D.P. Necrotizing soft tissue invasive aspergillosis in a cancer patient treated with immunosupressants due to checkpoint inhibitor-induced hepatitis. J. Infect. 2020, 80, 232–254. [Google Scholar] [CrossRef]
- Gupta, A.; Tun, A.; Ticona, K.; Baqui, A.; Guevara, E. Invasive Aspergillosis in a Patient with Stage III (or 3a or 3b) Non-Small-Cell Lung Cancer Treated with Durvalumab. Case Rep. Oncol. Med. 2019, 2019, 2178925-4. [Google Scholar] [CrossRef]
- Taima, K.; Tanaka, H.; Itoga, M.; Ishioka, Y.; Kurose, A.; Tasaka, S. Destroyed lung due to sustained inflammation after chemoradiotherapy followed by durvalumab. Respirol. Case Rep. 2020, 8, e00580. [Google Scholar] [CrossRef]
- Apostolopoulou, A.; Fishman, J.A. The Pathogenesis and Diagnosis of Pneumocystis jiroveci Pneumonia. J. Fungi 2022, 8, 1167. [Google Scholar] [CrossRef]
- Schwarz, M.; Kocher, F.; Niedersuess-Beke, D.; Rudzki, J.; Hochmair, M.; Widmann, G.; Hilbe, W.; Pircher, A. Immunosuppression for Immune Checkpoint-related Toxicity Can Cause Pneumocystis Jirovecii Pneumonia (PJP) in Non–small-cell Lung Cancer (NSCLC): A Report of 2 Cases. Clin. Lung Cancer 2018, 20, e247–e250. [Google Scholar] [CrossRef]
- Si, S.; Erickson, K.; Evageliou, N.; Silverman, M.; Kersun, L. An Usual Presentation of Pneumocystis jirovecii Pneumonia in a Woman Treated With Immune Checkpoint Inhibitor. J. Pediatr. Hematol. 2020, 43, e163–e164. [Google Scholar] [CrossRef] [PubMed]
- Vasconcellos, V.F.; Marta, G.N.; da Silva, E.M.; Gois, A.F.; de Castria, T.B.; Riera, R. Cisplatin versus carboplatin in combination with third-generation drugs for advanced non-small cell lung cancer. Cochrane Database Syst. Rev. 2020, 2020, CD009256. [Google Scholar] [CrossRef]
- Nakahama, K.; Tamiya, A.; Taniguchi, Y.; Sasaki, Y.; Akira, M.; Atagi, S. Severe acute interstitial lung disease after nivolumab in three non-small cell lung cancer patients with imaging findings of airway obstruction adjacent to lung tumors. J. Infect. Chemother. 2017, 23, 826–829. [Google Scholar] [CrossRef]
- Kim, T.; Kim, J. P3.CR-16 A Case of Toxic Hepatic Event Occurring in Combination Treatment with Nivolumab and Anti-Tuberculosis in Advanced Lung Cancer. J. Thorac. Oncol. 2018, 13, S1034. [Google Scholar] [CrossRef] [Green Version]
- Bukamur, H.; Katz, H.; Alsharedi, M.; Alkrekshi, A.; Shweihat, Y.R.; Munn, N.J. Immune Checkpoint Inhibitor-Related Pulmonary Toxicity: Focus on Nivolumab. South Med. J. 2020, 113, 600–605. [Google Scholar] [CrossRef] [PubMed]
- Maeno, K.; Fukuda, S.; Oguri, T.; Niimi, A. Nivolumab-induced asthma in a patient with non-small-cell lung cancer. Ann. Oncol. 2017, 28, 2891. [Google Scholar] [CrossRef] [PubMed]
- Goyal, P.; Moyers, J.T.; Elgohary, B.G.; Hammami, M.B. Case Report: Nivolumab-Induced Autoimmune Pancreatitis. J. Immunother. Precis. Oncol. 2021, 4, 208–211. [Google Scholar] [CrossRef] [PubMed]
- Zhou, P.; Zhang, J.; Tan, Z.; Tang, J.; Li, B.; Peng, H.; Zhou, R.; Zhou, C. Pembrolizumab Induced Grade 4 Skin irAEs with NonSmall Cell Lung Cancer: Case Report and Review of Literature. Ann. Hematol. Oncol. 2021, 8, 1381. [Google Scholar] [CrossRef]
- Mouri, A.; Kaira, K.; Shiono, A.; Miura, Y.; Kagamu, H. Severe Thrombocytopenia Associated with Pembrolizumab in Patients with Non-small Cell Lung Cancer (NSCLC): A Case Report and Literature Review. Vivo 2020, 34, 877–880. [Google Scholar] [CrossRef] [Green Version]
- Kunimasa, K.; Nishino, K.; Kimura, M.; Inoue, T.; Tamiya, M.; Kumagai, T.; Imamura, F. Pembrolizumab-induced acute thrombosis: A case report. Medicine 2018, 97, e10772. [Google Scholar] [CrossRef]
- Terasaki, K.; Ueno, A.; Mizuno, C.; Shima, T.; Okanoue, T. Gastrointestinal: Pembrolizumab-induced gastric ulcer occurring as an immune-related adverse event. J. Gastroenterol. Hepatol. 2022, 37, 1472. [Google Scholar] [CrossRef]
- Sonehara, K.; Tateishi, K.; Araki, T.; Komatsu, M.; Akahane, J.; Yamamoto, H.; Hanaoka, M. Pembrolizumab-Induced Adrenal Insufficiency in Patients with Untreated Advanced Non-Small Cell Lung Cancer: A Case Series. Case Rep. Oncol. 2021, 14, 1561–1566. [Google Scholar] [CrossRef] [PubMed]
- Inui, G.; Funaki, Y.; Makino, H.; Touge, H.; Arai, K.; Kuroda, K.; Hirayama, Y.; Kato, R.; Nonaka, T.; Yamane, K.; et al. Pembrolizumab-induced aseptic meningitis in a patient with non-small cell lung cancer: A case report and literature review of aseptic meningitis as an immune-related adverse event. Mol. Clin. Oncol. 2022, 17, 120. [Google Scholar] [CrossRef] [PubMed]
- Kichenadasse, G.; Miners, J.O.; Mangoni, A.A.; Rowland, A.; Hopkins, A.M.; Sorich, M.J. Multiorgan Immune-Related Adverse Events During Treatment with Atezolizumab. J. Natl. Compr. Cancer Netw. 2020, 18, 1191–1199. [Google Scholar] [CrossRef] [PubMed]
- Khadilkar, A.; Patel, K. S2686 A Rare Case of Atezolizumab-Induced Hepatotoxicity. Am. J. Gastroenterol. 2020, 115, S1406. [Google Scholar] [CrossRef]
- Nader, R.; Tannoury, E.; Rizk, T.; Ghanem, H.; Junior, E.L. Atezolizumab-induced encephalitis in a patient with metastatic breast cancer: A case report and review of neurological adverse events associated with checkpoint inhibitors. Autops. Case Rep. 2021, 11, e2021261. [Google Scholar] [CrossRef]
- Lacouture, M.E.; Wolchok, J.D.; Yosipovitch, G.; Kähler, K.C.; Busam, K.J.; Hauschild, A. Ipilimumab in patients with cancer and the management of dermatologic adverse events. J. Am. Acad. Dermatol. 2014, 71, 161–169. [Google Scholar] [CrossRef]
- Weber, J.S.; Kähler, K.C.; Hauschild, A. Management of Immune-Related Adverse Events and Kinetics of Response with Ipilimumab. J. Clin. Oncol. 2012, 30, 2691–2697. [Google Scholar] [CrossRef]
- Tarhini, A. Immune-Mediated Adverse Events Associated with Ipilimumab CTLA-4 Blockade Therapy: The Underlying Mechanisms and Clinical Management. Scientifica 2013, 2013, 857519. [Google Scholar] [CrossRef]
- Kotwal, A.; Kottschade, L.; Ryder, M. PD-L1 Inhibitor-Induced Thyroiditis Is Associated with Better Overall Survival in Cancer Patients. Thyroid 2020, 30, 177–184. [Google Scholar] [CrossRef]
- Walsh, R.J.; Sundar, R.; Lim, J.S.J. Immune checkpoint inhibitor combinations—Current and emerging strategies. Br. J. Cancer 2023, 128, 1415–1417. [Google Scholar] [CrossRef]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
- Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
- Naidoo, J.; Page, D.B.; Li, B.T.; Connell, L.C.; Schindler, K.; Lacouture, M.E.; Postow, M.A.; Wolchok, J.D. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann. Oncol. 2015, 26, 2375–2391. [Google Scholar] [CrossRef]
- Puzanov, I.; Diab, A.; Abdallah, K.; Bingham, C.O.; Brogdon, C.; Dadu, R.; Hamad, L.; Kim, S.; Lacouture, M.E.; LeBoeuf, N.R.; et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Man-agement Working Group. J. Immunother. Cancer 2017, 5, 95. [Google Scholar] [CrossRef] [Green Version]
- Adam, K.; Iuga, A.; Tocheva, A.S.; Mor, A. A novel mouse model for checkpoint inhibitor-induced adverse events. PLoS ONE 2021, 16, e0246168. [Google Scholar] [CrossRef] [PubMed]
- Nishino, M.; Sholl, L.M.; Hodi, F.S. Anti–PD-1–Related Pneumonitis during Cancer Immunotherapy. N. Engl. J. Med. 2015, 373, 286–288. [Google Scholar] [CrossRef] [Green Version]
- Akbari, O.; Stock, P.; Singh, A.K.; Lombardi, V.; Lee, W.-L.; Freeman, G.J.; Sharpe, A.H.; Umetsu, D.T.; Dekruyff, R.H. PD-L1 and PD-L2 modulate airway inflammation and iNKT-cell-dependent airway hyperreactivity in opposing directions. Mucosal Immunol. 2010, 3, 81–91. [Google Scholar] [CrossRef] [Green Version]
- Downey, S.G.; Klapper, J.A.; Smith, F.O.; Yang, J.C.; Sherry, R.M.; Royal, R.E.; Kammula, U.S.; Hughes, M.S.; Allen, T.E.; Levy, C.L.; et al. Prognostic Factors Related to Clinical Response in Patients with Metastatic Melanoma Treated by CTL-Associated Antigen-4 Blockade. Clin. Cancer Res. 2007, 13, 6681–6688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matsuya, T.; Nakamura, Y.; Matsushita, S.; Tanaka, R.; Teramoto, Y.; Asami, Y.; Uehara, J.; Aoki, M.; Yamamura, K.; Nakamura, Y.; et al. Vitiligo expansion and extent correlate with durable response in anti-programmed death 1 antibody treatment for advanced melanoma: A multi-institutional retrospec-tive study. J. Dermatol. 2020, 47, 629–635. [Google Scholar] [CrossRef]
- Ma, B.; Anandasabapathy, N. Immune Checkpoint Blockade and Skin Toxicity Pathogenesis. J. Investig. Dermatol. 2021, 142, 951–959. [Google Scholar] [CrossRef] [PubMed]
- Sibaud, V. Dermatologic Reactions to Immune Checkpoint Inhibitors: Skin Toxicities and Immunotherapy. Am. J. Clin. Dermatol. 2018, 19, 345–361. [Google Scholar] [CrossRef]
- Bonigen, J.; Raynaud-Donzel, C.; Hureaux, J.; Kramkimel, N.; Blom, A.; Jeudy, G.; Breton, A.-L.; Hubiche, T.; Bedane, C.; Legoupil, D.; et al. Anti-PD1-induced psoriasis: A study of 21 patients. J. Eur. Acad. Dermatol. Venereol. 2017, 31, e254–e257. [Google Scholar] [CrossRef]
- Nikolaou, V.; Sibaud, V.; Fattore, D.; Sollena, P.; Ortiz-Brugués, A.; Giacchero, D.; Romano, M.C.; Riganti, J.; Lallas, K.; Peris, K.; et al. Immune checkpoint-mediated psoriasis: A multicenter European study of 115 patients from the European Network for Cutaneous Adverse Event to Oncologic Drugs (ENCADO) group. J. Am. Acad. Dermatol. 2020, 84, 1310–1320. [Google Scholar] [CrossRef]
- Postow, M.A. Managing Immune Checkpoint-Blocking Antibody Side Effects. Am. Soc. Clin. Oncol. Educ. Book 2015, 35, 76–83. [Google Scholar] [CrossRef]
- Belum, V.R.; Benhuri, B.; Postow, M.A.; Hellmann, M.D.; Lesokhin, A.M.; Segal, N.H.; Motzer, R.J.; Wu, S.; Busam, K.J.; Wolchok, J.D.; et al. Characterization and management of dermatologic adverse events to agents targeting the PD-1 receptor. Eur. J. Cancer 2016, 60, 12–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hua, C.; Boussemart, L.; Mateus, C.; Routier, E.; Boutros, C.; Cazenave, H.; Viollet, R.; Thomas, M.; Roy, S.; Benannoune, N.; et al. Association of Vitiligo with Tumor Response in Patients with Metastatic Melanoma Treated with Pembrolizumab. JAMA Dermatol. 2016, 152, 45–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berner, F.; Bomze, D.; Diem, S.; Ali, O.H.; Fässler, M.; Ring, S.; Niederer, R.; Ackermann, C.J.; Baumgaertner, P.; Pikor, N.; et al. Association of Checkpoint Inhibitor–Induced Toxic Effects with Shared Cancer and Tissue Antigens in Non-Small Cell Lung Cancer. JAMA Oncol. 2019, 5, 1043–1047. [Google Scholar] [CrossRef]
- Maker, A.V.; Attia, P.; Rosenberg, S.A. Analysis of the Cellular Mechanism of Antitumor Responses and Autoimmunity in Pa-tients Treated with CTLA-4 Blockade. J. Immunol. 2005, 175, 7746–7754. [Google Scholar] [CrossRef]
- Dulos, J.; Carven, G.J.; Van Boxtel, S.J.; Evers, S.; Driessen-Engels, L.J.A.; Hobo, W.; Gorecka, M.A.; de Haan, A.F.J.; Mulders, P.; Punt, C.J.A.; et al. PD-1 blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from patients with prostate and advanced melanoma cancer. J. Immunother. 2012, 35, 169–178. [Google Scholar] [CrossRef]
- Zak, K.M.; Grudnik, P.; Guzik, K.; Zieba, B.J.; Musielak, B.; Dömling, A.; Dubin, G.; Holak, T.A. Structural basis for small molecule targeting of the programmed death ligand 1 (PD-L1). Oncotarget 2016, 7, 30323–30335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tartof, S.Y.; Slezak, J.M.; Fischer, H.; Hong, V.; Ackerson, B.K.; Ranasinghe, O.N.; Frankland, T.B.; Ogun, O.A.; Zamparo, J.M.; Gray, S.; et al. Effectiveness of mRNA BNT162b2 COVID-19 vaccine up to 6 months in a large integrated health system in the USA: A retrospective cohort study. Lancet 2021, 398, 1407–1416. [Google Scholar] [CrossRef] [PubMed]
- Szabó, G.T.; Mahiny, A.J.; Vlatkovic, I. COVID-19 mRNA vaccines: Platforms and current developments. Mol. Ther. 2022, 30, 1850–1868. [Google Scholar] [CrossRef] [PubMed]
- Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef] [Green Version]
- Ordikhani, F.; Uehara, M.; Kasinath, V.; Dai, L.; Eskandari, S.K.; Bahmani, B.; Yonar, M.; Azzi, J.R.; Haik, Y.; Sage, P.T.; et al. Targeting antigen-presenting cells by anti–PD-1 nanoparticles augments antitumor immunity. J. Clin. Investig. 2018, 3, e122700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allen, S.D.; Liu, X.; Jiang, J.; Liao, Y.-P.; Chang, C.H.; Nel, A.E.; Meng, H. Immune checkpoint inhibition in syngeneic mouse cancer models by a silicasome nanocarrier delivering a GSK3 inhibitor. Biomaterials 2021, 269, 120635. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Gao, D.; Zhao, J.; Yang, G.; Guo, M.; Wang, Y.; Ren, X.; Kim, J.S.; Jin, L.; Tian, Z.; et al. Thermal immuno-nanomedicine in cancer. Nat. Rev. Clin. Oncol. 2023, 20, 116–134. [Google Scholar] [CrossRef]
Drug | Target | Organ Damage Induced by ICI | Refs. |
---|---|---|---|
Nivolumab | PD-1 | Hepatotoxicity, ILD, rash, pruritus, asthma, pancreatitis. | [6,83,84,85,86,87] |
Pembrolizumab | PD-1 | Meningitis, thrombosis, gastric ulcer, thrombocytopenia, adrenal insufficiency, cutaneous erythema. | [88,89,90,91,92,93] |
Atezolizumab | PD-L1 | Skin rash, pneumonitis, hepatotoxicity, peripheral neuropathy, encephalitis. | [94,95,96] |
Ipilimumab | CTLA-4 | Hepatitis, rash, pruritus, colitis, thyroiditis. | [97,98,99] |
Avelumab | PD-L1 | Thyroiditis, rash and pruritus, hepatitis. | [54,100] |
Durvalumab | PD-L1 | Lung damage | [78] |
Dostarlimab | PD-1 | Fatigue, diarrhoea and nausea, neutropenia | [59,60] |
Relatlimab | LAG-3 | Increased levels of lipase, acute pulmonary oedema, and pneumonitis | [9] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ocaña-Guzmán, R.; Osorio-Pérez, D.; Chavez-Galan, L. Opportunistic Infections and Immune-Related Adverse Events Associated with Administering Immune Checkpoint Inhibitors: A Narrative Review. Pharmaceuticals 2023, 16, 1119. https://doi.org/10.3390/ph16081119
Ocaña-Guzmán R, Osorio-Pérez D, Chavez-Galan L. Opportunistic Infections and Immune-Related Adverse Events Associated with Administering Immune Checkpoint Inhibitors: A Narrative Review. Pharmaceuticals. 2023; 16(8):1119. https://doi.org/10.3390/ph16081119
Chicago/Turabian StyleOcaña-Guzmán, Ranferi, Diego Osorio-Pérez, and Leslie Chavez-Galan. 2023. "Opportunistic Infections and Immune-Related Adverse Events Associated with Administering Immune Checkpoint Inhibitors: A Narrative Review" Pharmaceuticals 16, no. 8: 1119. https://doi.org/10.3390/ph16081119