1,275
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Endocrine toxicity of immune checkpoint inhibitors: a network meta-analysis of the current evidence

      Published
      review-article
      Bookmark

            Abstract

            Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment but pose a challenge of immune-related adverse events (irAEs), particularly endocrine toxicity, that can severely compromise patient well-being. Existing research has often been limited in scope and has not provided comprehensive safety profiles across the diverse range of ICI therapies. We addressed this gap by performing a network meta-analysis on 55 randomized controlled trials involving 32,522 patients. Using STATA to calculate the surface under the cumulative ranking curve, we ranked the safety of various ICI monotherapies and combination therapies. ICIs were found to increase the risk of endocrine toxicities, such as hypothyroidism, hyperthyroidism, hypophysitis, thyroiditis, and adrenal insufficiency; this risk was greater with dual ICI regimens. Specifically, cytotoxic T lymphocyte associated antigen-4 (CTLA-4) inhibitors, such as ipilimumab, are closely associated with hypophysitis, whereas programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) inhibitors, notably pembrolizumab and nivolumab, predispose patients to thyroid-related dysfunction, such as hyperthyroidism, hypothyroidism, and thyroiditis. Interestingly, nivolumab showed no elevated risk of adrenal dysfunction, in contrast to the elevated risk observed with other ICI treatments. This study provides critical evidence-based insights for optimizing the risk-benefit balance of ICI therapies in clinical practice.

            Main article text

            1. INTRODUCTION

            ICIs have led to a groundbreaking shift in cancer therapy, by significantly improving treatment outcomes across various malignancies [1, 2]. These treatments have been approved for various cancers, including malignant melanoma, non-small cell lung cancer (NSCLC), and renal cell carcinoma (RCC), each of which shows a distinct epidemiological pattern and response to ICIs [3]. The integration of ICIs is addressing critical needs in oncology, by targeting CTLA-4, PD-1, and PD-L1. Research has underscored the efficacy of CTLA-4 inhibitors in melanoma treatment, through significantly increasing survival rates [4]. Similarly, PD-1/PD-L1 inhibitors have revolutionized lung cancer treatment by enabling the development of personalized strategies through the analysis of PD-1 and PD-L1 expression levels, and enhancing the precision of immunotherapy [5].

            ICIs have also shown promise in treating other cancers, such as triple-negative breast cancer and HER2-positive breast cancer with PD-1 inhibitors, and in improving overall survival in platinum-resistant head and neck squamous cell carcinoma with nivolumab [6, 7]. For patients with certain types of metastatic RCC, a combination of nivolumab and ipilimumab is now the preferred initial treatment, taking into account patient tolerance and quality of life [8]. However, despite their broad efficacy, ICIs exhibit unique response patterns and a range of tumor responses that differ from those with traditional therapies, such as chemotherapy, radiotherapy, and targeted therapy. Challenges include delayed responses, pseudo-progression, hyper-progression, and immune-related adverse events (irAEs), which can cause severe endocrine toxicity affecting the thyroid, adrenal glands, and pancreas [9, 10]. These complications may significantly affect patient well-being and survival, thus underscoring the need for careful patient monitoring and a deeper understanding of the epidemiological data associated with ICIs and cancer treatment.

            In recent years, emerging treatment strategies based on ICIs, such as combinations with chemotherapy, anti-angiogenic drugs, and dual ICI therapies, have entered clinical practice [11]. Extensive clinical research evaluating these combination therapies has demonstrated that both chemotherapy with ICIs and dual ICI treatments substantially improve patient outcomes and prognosis [1214]. Notably, dual ICI treatments, particularly ipilimumab and nivolumab, have shown objective response rates of 58–71% in phase III clinical trials for metastatic clear cell renal cell carcinoma [15, 16]. The enhanced therapeutic effects are attributed to the concurrent blockade of CTLA-4 and PD-1/PD-L1 pathways, thereby fostering synergistic anti-cancer effects. Nonetheless, although these combination therapies may offer synergistic benefits, they can also significantly increase toxicity levels [17]. This concern is particularly relevant to ICI-induced endocrine toxicity: focused research has extensively explored this toxicity, yet current studies have often been constrained by their narrow scope and methods, thus limiting their applicability to the diverse and evolving array of ICI treatments. This limitation is particularly salient in the comparison of safety profiles across multiple ICI strategies—a task for which traditional meta-analysis is often inadequate. To address this gap, this study leverages the sophisticated capabilities of network meta-analysis (NMA), an analytical approach uniquely suited for comparing multiple interventions in an integrated manner. Unlike traditional meta-analyses, NMA enables the comparison of multiple treatments both directly and indirectly, thus offering a comprehensive safety ranking for specific endocrine dysfunctions across a broad range of ICI therapies. The use of NMA methods represents an important advance over previous studies, by enabling a more nuanced understanding of ICI-related endocrine toxicity.

            This study was aimed at providing a robust, quantitative safety ranking of endocrine disruptions induced by various ICIs, in both monotherapy and combination therapy settings, to provide a valuable resource for clinicians. Our comprehensive analysis not only augments current understanding of ICI-induced endocrine toxicity but also provides a critical decision-making tool for clinicians aiming to personalize ICI regimens while minimizing associated risks.

            2. METHODS

            2.1 Search strategy

            We conducted a literature search of PubMed, Embase, the Cochrane Library, and ClinicalTrials.gov to identify relevant studies published in English through December 2022. Our search strategy used the following keywords: ipilimumab, tremelimumab, pembrolizumab, nivolumab, camrelizumab, atezolizumab, durvalumab, avelumab, CTLA-4, PD-1, PD-L1, and immune checkpoint inhibitor. The search strategy is detailed in the Table S1 .

            2.2 Inclusion and exclusion criteria

            To select eligible studies, we followed the patient population, interventions, controls, and outcome indicators principles of evidence-based medicine, applying specific inclusion and exclusion criteria. We included randomized controlled trials (RCTs) of ICI monotherapy or combination therapy that reported data on immune-related endocrine toxicity, such as hyperthyroidism, hypothyroidism, hypophysitis, adrenal insufficiency, and thyroiditis. When multiple studies reported data on the same trial, we included only the most complete and updated data. Two investigators independently screened all titles, abstracts, and full texts for eligibility, and any discrepancies were resolved through consultation. In this process, we excluded non-English articles, as well as reviews, case reports, editorials, meta-analyses, letters to the editor, and conference records.

            2.3 Data extraction

            We extracted the following information from the included studies: clinical trial NCT number; year of study; trial location; trial phase; type of cancer; total number of patients; treatment regimen; median follow-up time; number of patients experiencing immune-related hyperthyroidism, hypothyroidism, hypophysitis, adrenal insufficiency, or thyroiditis; and total sample size. To ensure consistency and accuracy, we extracted research data and detailed content by using standardized electronic spreadsheets.

            2.4 Quality assessment

            We used the Cochrane Risk of Bias Assessment Tool to evaluate the potential risk of bias in these clinical trials. This tool evaluates the quality of clinical studies in seven dimensions: random sequence generation (selection bias), allocation concealment (selection bias), blinding to intervention (performance bias), blinding to outcome assessment (detection bias), incomplete outcome data (attrition bias), selective reporting (reporting bias), and other biases. Study quality was classified as low risk of bias (+), high risk of bias (-), or unclear (?).

            2.5 Statistical analysis

            We used Stata 16.0 software to perform NMA and generate evidence network diagrams for the various ICI treatment regimens. Binary variables and odds ratios (OR) with 95% confidence intervals (CI) were used to measure outcomes, and P values less than 0.05 were considered significant. We assessed heterogeneity among studies by using direct meta-analysis and the I2 test, selecting fixed-effects or random-effects models accordingly. If significant heterogeneity was present, with I2 ≥50%, a great influence on the statistical results was indicated, and a random-effect model was used; otherwise, a fixed-effect model was used. For closed-loop studies, we used OR and 95% CI for the heterogeneity test, and used a fixed-effects model if the value contained 0, and P was greater than 0.05. We evaluated consistency and inconsistency with the node-splitting method, with a P value greater than 0.05 indicating good consistency. We estimated overall treatment rankings with the surface under the cumulative ranking (SUCRA) and a league table for each endocrine-related adverse effect. We used funnel plots to test for bias of the included studies.

            3. RESULTS

            3.1 Literature selection process and characteristics of the included literature

            Figure 1 illustrates the flowchart of the study selection process. Initially, we identified 2,942 potentially relevant studies through electronic searches of PubMed, Embase, the Cochrane Library, and ClinicalTrials.gov. After screening the titles and abstracts, we assessed 406 potentially eligible articles for eligibility. Ultimately, we included 55 head-to-head RCTs comprising 32,522 patients for the quantitative NMA synthesis. Eleven phase II trials and 44 phase III RCTs were among those included. We analyzed data on endocrine toxicity from 48 studies on hypothyroidism, 26 studies on hyperthyroidism, 16 studies on hypophysitis, 15 studies on adrenal insufficiency, and 11 studies on thyroiditis. The detailed baseline characteristics of the included studies are presented in Table 1 .

            Figure 1 |

            Flow diagram illustrating the literature selection process for the NMA.

            Table 1 |

            Baseline characteristics of 55 randomized controlled trials for network meta-analysis by cancer type.

            Study IDAuthorYearRegionPhaseCancer typeTotal No.Safety analysis No.ArmTreatmentNCT No.
            KEYNOTE 006 [18]Robert2015MNIIIMM8342781PEM 10 mg/kg every 2 weeks01866319
            2772PEM 10 mg/kg every 3 weeks
            2563IPI 3 mg/kg every 3 weeks
            OAK [19]Rittmeyer2017JapanIIINSCLC101561ATE 1,200 mg every 3 weeks02008227
            452DTX 75 mg/m2 every 3 weeks
            CheckMate 037 [20]Larkin2017MNIIIMM4052681NIV 3 mg/kg every 2 weeks01721746
            1022DAC 1,000 mg/m2 or CBP AUC = 6 + PTX 175 mg/m2 every 3 weeks
            CheckMate 025 [21]Motzer2015MNIIIRCC8214061NIV 3 mg/kg every 2 weeks01668784
            3972EVE 10 mg orally once daily
            KEYNOTE 045 [22]Bellmunt2017MNIIIUC5422661PEM 200 mg every 3 weeks02256436
            2552PTX, DTX, or VIN
            CA184-162 [23]Bang2017MNIIIGC/GEJC114571IPI 10 mg/kg every 3 weeks01585987
            452Best supportive care
            CA184-169 [24]Ascierto2017MNIIIMM7273641IPI 10 mg/kg every 3 weeks01515189
            3622IPI 3 mg/kg every 3 weeks
            KEYNOTE 024 [25]Reck2016MNIIINSCLC3051541PEM 200 mg every 3 weeks02142738
            1502PT-based chemotherapy
            KEYNOTE021 [26]Langer2016USA, TaiwanIINSCLC123591PEM 200 mg + CBP (AUC = 5) + PMT 500 mg/m2 every 3 weeks02039674
            622CBP (AUC = 5) + PMT 500 mg/m2 every 3 weeks
            CheckMate 069 [27]Hodi2016USA, FranceIIIMM142941NIV 1 mg/kg + IPI 3 mg/kg every 3 weeks01927419
            462IPI 3 mg/kg every 3 weeks
            KEYNOTE 010 [28]Herbst2016MNII/IIINSCLC1,0343391PEM 2 mg/kg every 3 weeks01905657
            3432PEM 10 mg/kg every 3 weeks
            3093DTX 75 mg/m2 every 3 weeks
            POPLAR [29]Fehrenbacher2016MNIINSCLC2871421ATE 1,200 mg every 3 weeks01903993
            1352DTX 75 mg/m2 every 3 weeks
            KEYNOTE 002 [30]Ribas2015MNIIMM5401781PEM 2 mg/kg every 3 weeks01704287
            1792PEM 10 mg/kg every 3 weeks
            1713ICC (PTX and CBP, PTX, CBP, DTIC oral TEM)
            A3671009 [31]Ribas2013MNIIIMM6553251TRE 15 mg/kg every 90 days00257205
            3162TEM 200 mg/m2 every 4 weeks or DTIC 1,000 mg/m2 every 3 weeks
            JAVELIN Bladder 100 [32]Powles2020IIIUC6893441AVE 10 mg/kg every 2 weeks02603432
            3452Best supportive care
            JAVELIN Renal 101 [33]Motzer2019MNIIIRCC8864421AVE 10 mg/kg every 2 weeks + AXI 5 mg twice daily02684006
            4442SUN 50 mg orally once daily
            CheckMate 067 [34]Hodi2018MNIIIMM9373131NIV 1 mg/kg + IPI 3 mg/kg every 3 weeks01844505
            3132NIV 3 mg/kg every 2 weeks
            3113IPI 3 mg/kg every 3 weeks
            CheckMate 511 [35]Lebbe2019MNIIIMM3581801NIV 3 mg/kg + IPI 1 mg/kg every 3 weeks02714218
            1782NIV 1 mg/kg + IPI 3 mg/kg every 3 weeks
            NRG GY003 [36]Zamarin2020USAIIOC100491NIV 3 mg/kg every 2 weeks02498600
            512NIV 3 mg/kg + IPI 1 mg/kg every 3 weeks
            ANNONC 152 [37]Ferris2020MNIIIHNSC274781DUR 10 mg/kg every 2 weeks02369874
            1042DUR 20 mg/kg + TRE 1 mg/kg every 4 weeks
            923SOC (CET, taxane, MTX or FLU)
            KEYNOTE 252 [38]Long2019MNIIIMM7063531PEM 200 mg every 3 weeks + epacadostat 100 mg twice daily02752074
            3522PEM 200 mg every 3 weeks
            OpACIN neo [39]Rozeman2019MNIIMM89301IPI 3 mg/kg + NIV 1 mg/kg every 3 weeks02977052
            302IPI 1 mg/kg + NIV 3 mg/kg every 3 weeks
            263IPI 3 mg/kg every 3 weeks
            KEYNOTE 407 [40]Paz-Ares2018MNIIINSCLC5592781PEM 200 mg every 3 weeks + CBP (AUC = 6) + PTX 200 mg/m2 or Nab-PTX 100 mg/m2 02775435
            2802CBP (AUC = 6) + PTX 200 mg/m2 or Nab-PTX 100 mg/m2
            KEYNOTE 042 [41]Mok2019MNIIINSCLC1,2746361PEM 200 mg every 3 weeks02220894
            6152ICC (PT-based chemotherapy)
            KEYNOTE 048 [42]Burtness2019MNIIIHNSC8823001PEM 200 mg every 3 weeks02358031
            2762PEM 200 mg every 3 weeks + CBP (AUC = 5) or CIS 100 mg/m2 and 5-FLU 1,000 mg/m2 per day for 4 consecutive days
            2873CET 400 mg/m2 loading dose, then 250 mg/m2 per week + CBP (AUC = 5) or CIS 100 mg/m2 and 5-FLU 1,000 mg/m2 per day for 4 consecutive days
            KEYNOTE 189 [43]Gadgeel2020MNIIINSCLC6164051PEM 200 mg every 3 weeks + PMT 500 mg/m2 every 3 weeks + CBP (AUC = 5) or CIS 75 mg/m2 every 3 weeks02578680
            2022PMT 500 mg/m2 every 3 weeks + CBP (AUC = 5) or CIS 75 mg/m2 every 3 weeks
            KEYNOTE 522 [44]Schmid2022MNIIIBC1,1747831PEM 200 mg every 3 weeks + PTX 80 mg/m2 + CBP 5 mg/m2 03036488
            3892PTX 80 mg/m2 + CBP 5 mg/m2
            KEYNOTE 024 [45]Reck2019MNIIINSCLC3051541PEM 200 mg every 3 weeks02142738
            1502PT-based chemotherapy
            IMpassion130 [46]Schmid2019MNIIIBC9024531ATE 840 mg days 1 and 15 of each 28-day cycle + Nab-PTX 100 mg/m2 days 1, 8, and 15 of each 28-day cycle02425891
            4372Nab-PTX 100 mg/m2 days 1, 8, and 15 of each 28-day cycle
            KEYNOTE 177 [47]André2020MNIIICRC3071531PEM 200 mg every 3 weeks02563002
            1432mFOLFOX6 or FOLFIRI + BEV 5 mg/kg every 2 weeks or CET 400 mg/m2 over 2 hours, then 250 mg/m2 over 1 hour weekly in each 2-week cycle
            CheckMate 214 [48]Motzer2019MNIIIRCC1,0965471NIV 3 mg/kg + IPI 1 mg/kg every 3 weeks for four doses, followed by NIV3 mg/kg every 2 weeks02231749
            5352SUN 50 mg orally once daily for 4 weeks
            KEYNOTE 040 [49]Cohen2018MNIIIHNSC4802461PEM 200 mg every 3 weeks02252042
            2342MTX 40 mg/m2 days 1, 8, and 15 of each 3-week cycle, DTX 75 mg/m2 day 1 of each 3-week cycle or CET 400 mg/m2 loading dose on day 1 and 250 mg/m2 on days 8 and 15 of cycle 1, followed by CET 250 mg/m2 on days 1, 8, and 15 of each subsequent 3-week cycle
            IMmotion 151 [50]Rini2019MNIIIRCC9154541ATE 1,200 mg every 3 weeks + BEV 15 mg/kg every 3 weeks02420821
            4612SUN 50 mg orally once daily for 4 weeks
            KEYNOTE 045 [51]Bellmunt2017MNIIIUC5422661PEM 200 mg every 3 weeks02256436
            2552PTX 175 mg/m2, DTX 75 mg/m2, or VIN 320 mg/m2 every 3 weeks
            KEYNOTE 062 [52]Shitara2020MNIIIGC7632541PEM 200 mg every 3 weeks02494583
            2502PEM 200 mg every 3 weeks + CIS 80 mg/m2 + FLU 800 mg/m2/d day 1 to 5 or CAP 1,000 mg/m2 twice daily day 1–14 every 3 weeks
            2443CIS 80 mg/m2/d on day 1 + FLU 800 mg/m2/d on days 1–5 or CAP 1,000 mg/m2 twice daily
            IMspire 150 [53]Gutzmer2020MNIIIMM7772301ATE 840 mg + VEM 960 mg twice per day + COM 60 mg once daily02908672
            2812VEM 960 mg twice per day + COM 60 mg once daily
            IMpassion 031 [54]Mittendorf2020MNIIIBC4551641ATE 840 mg + Nab-PTX 125 mg/m2 every week + DOX 60 mg/m2 and CTX 600 mg/m2 every 2 weeks03197935
            1672Nab-PTX 125 mg/m2 every week, followed by DOX 60 mg/m2 and CTX 600 mg/m2 every 2 weeks
            CheckMate 227 [55]Hellmann2018MNIIINSCLC1,1893961NIV 3 mg/kg every 2 weeks + IPI 1 mg/kg every 6 weeks02477826
            3962NIV 240 mg every 2 weeks
            3973PT-based chemotherapy
            CheckMate 9 LA [56]Paz-Ares2021MNIIINSCLC1,1503581NIV 360 mg every 3 weeks + IPI 1 mg/kg every 6 weeks + PT-based chemotherapy03215706
            3492PT-based chemotherapy
            KEYNOTE426 [57]Powles2020MNIIIRCC8614291PEM 200 mg every 3 weeks + conventional therapy02853331
            4252SUN 50 mg once daily
            KEYNOTE 604 [58]Rudin2020MNIIISCLC4532231PEM 200 mg every 3 weeks + VP16 100 mg/m2 + (AUC = 5) or CIS 75 mg/m2 03066778
            2232VP16 100 mg/m2 + (AUC = 5)or CIS 75 mg/m2
            CheckMate 066 [59]Ascierto2018MNIIIMM4182061NIV 3 mg/kg every 2 weeks01721772
            2052DAC 1,000 mg/m2 every 3 weeks
            I-SPY2 Trial [60]Nanda2020MNIIBC250691PEM 200 mg every 3 weeks + PTX 80 mg/m2 every week + DOX 60 mg/m2 + CTX 600 mg/m2 every 2 to 3 weeks01042379
            1812PTX 80 mg/m2 every week + DOX 60 mg/m2 + CTX 600 mg/m2 every 2–3 weeks
            CheckMate 238 [61]Ascierto2020MNIIIMM9054521NIV 3 mg/kg every 2 weeks02388906
            4532IPI 10 mg/kg every 3 weeks
            JAVELIN Gastric 300 [62]Bang2018MNIIIGC3711841AVE 10 mg/kg every 2 weeks02625623
            1772PTX 80 mg/m2 or IRI 150 mg/m2 every week
            CASPIAN [63]Goldman2020MNIIISCLC9722661DUR 1,500 mg + TRE 75 mg every 3 weeks + PT + VP16 80–100 mg/m2 03043872
            2652DUR 1,500 mg every 3 weeks + PT + VP16
            2663PT + VP16
            IMblaze 370 [64]Eng2019MNIIICRC3631831ATE 840 mg + COM 60 mg once daily02788279
            902ATE 1,200 mg every 3 weeks
            903REG 160 mg once daily
            E1609 [65]Ahmad2019MNIIIMM1,0185161IPI 3 mg/kg every 3 weeks01274338
            5032IPI 10 mg/kg every 3 weeks
            Jing Nie et al. [66]Nie2019ChinaIIHL86191CAM 200 mg every 3 weeks03250962
            672CAM 200 mg every 3 weeks + DTB 10 mg/d day 1–5
            PROLUNG [67]Arrieta2020MexicoIINSCLC78401PEM 200 mg every 3 weeks + DTX 75 mg/m2 every 3 weeks02574598
            382DTX 75 mg/m2 every 3 weeks
            KATE2 [68]Emens2020MNIIBC2001321ATE 1,200 mg + TDM-1 3.6 mg/kg02924883
            682Trastuzumab emtansine 3.6 mg/kg
            CameL [69]Zhou2020ChinaIIINSCLC4192051CAM 200 mg every 3 weeks + CBP (AUC = 5) + PMT 500 mg/m2 every 3 weeks03134872
            2072CBP (AUC = 5) + PMT 500 mg/m2 every 3 weeks
            ESCORT [70]Huang2020ChinaEC4572281CAM 200 mg every 2 weeks03099382
            2202DTX 75 mg/m2 every 3 weeks or IRI 180 mg/m2 every 2 weeks
            CheckMate 9ER [71]Choueiri2021MNIIIRCC6513231NIV 240 mg every 2 weeks + CZT 40 mg once daily03141177
            3282SUN 50 mg once daily
            KEYNOTE 181 [72]Kojima2020MNIIIEC6283141PEM 200 mg every 3 weeks02559687
            2962PTX 80–100 mg/m2 every week or DTX 75 mg/m2 every 3 weeks or IRI 180 mg/m2 every 2 weeks

            CTCAE = Common Terminology Criteria for Adverse Events; TrAE = treatment-related adverse event; MN = multinational; PD-L1 = programmed cell death ligand 1; MM = melanoma; NIV = nivolumab; IPI = ipilimumab; DAC = dacarbazine; PEM = pembrolizumab; NR = not reported; ICC = investigator’s choice chemotherapy; CBP = carboplatin; AUC = area under the curve; PT = platinum; PTX = paclitaxel; TEM = temozolomide; ATE = atezolizumab; CIS = cisplatin; GEM = gemcitabine; ETO = VP16; DTX = docetaxel; CR = concurrent regimen; PR = phased regimen; MTX = methotrexate; CET = cetuximab; EVE = everolimus; VIN = vinflunine; NSCLC = non-small cell lung cancer; RCC = renal cell carcinoma; GC = gastric carcinoma; GEJC = gastro-esophageal junction; UC = urothelial cancer; PMT = pemetrexed; AVE = avelumab; AXI = axitinib; SUN = sunitinib; OC = ovarian carcinoma; TRE = tremelimumab; SOC = standard of care; HNSC = head and neck squamous carcinoma; FLU = fluorouracil; BC = breast carcinoma; BEV = bevacizumab; CRC = colorectal carcinoma; CAP = capecitabine; VEM = vemurafenib; COM = cobimetinib; CTX = cyclophosphamide; DOX = doxorubicin; SCLC = small cell lung cancer; REG = regorafenib; HL = Hodgkin lymphoma; CAM = camrelizumab; DTB = decitabine; CZT = cabozanitinib; EC = esophageal carcinoma; IRI = irinotecan; DUR = durvalumab.

            3.2 Quality assessment results of the included studies

            Among the 55 studies included, 69.1% met the criteria for random sequence generation, 47.3% reported allocation concealment, 43.7% implemented blinding to the intervention, 96.4% implemented blinding to the outcome assessment, and 90.9% had complete outcome data. Furthermore, as shown in Figure S1 , selective reporting bias was not detected in 98.2% of the included studies. These results suggested that the studies included in this meta-analysis are of relatively high quality.

            3.3 Network meta-analysis results
            3.3.1 Network plot

            Figure 2 displays the network plots for five immune related-endocrine toxicity events: hypothyroidism, hyperthyroidism, hypophysitis, adrenal insufficiency, and thyroiditis. The 11 ICI treatment strategies in the analysis included four PD-1 inhibitors, three PD-L1 inhibitors, and two CTLA-4 inhibitors. The ICI monotherapy options included pembrolizumab (n = 2,675), ipilimumab (n = 1,809), nivolumab (n = 1,411), avelumab (n = 528), tremelimumab (n = 325), camrelizumab (307), atezolizumab (n = 288), and durvalumab (n = 78). ICI combination therapy included one ICI plus conventional therapy (n = 6,078), two ICIs plus conventional therapy (n = 624), and two ICIs (n = 2,043). In addition, 10,786 patients received conventional therapy.

            Figure 2 |

            Network plots for hypothyroidism (A), hyperthyroidism (B), hypophysitis (C), adrenal insufficiency (D), and thyroiditis (E) caused by different modes of drug administration, and patient number (F) included in different ICI regimens.

            3.3.2 Inconsistency testing

            The global inconsistency test revealed no significant heterogeneity for hypothyroidism, hyperthyroidism, hypophysitis, adrenal insufficiency, and thyroiditis ( Table S2 ), with all P values greater than 0.05. The I2 test showed high heterogeneity in hypothyroidism, hyperthyroidism, hypophysitis, and adrenal insufficiency, with I2 values of 85.7%, 75.4%, 73.4%, and 70.5%, respectively ( Figures S2–S6 ). The closed-loop inconsistency test, which assesses inconsistency under both direct and indirect comparisons, also demonstrated no significant inconsistency, as the 95% CI for all adverse reactions included 0 ( Tables S3–S7 ). The node-splitting method was used to test the overall inconsistency for each direct and indirect comparison, and all P values were greater than 0.05, indicating no significant inconsistency ( Tables S8–S12 ). Therefore, a consistency model was chosen for the NMA.

            3.3.3 League table and cumulative probability ranking
            3.3.3.1 Hypothyroidism

            ICI combination therapies, including one ICI plus conventional therapy, dual ICIs plus conventional therapy, and dual ICIs, were associated with a higher incidence of hypothyroidism than conventional therapy ( Figure 3a ). Treatment with dual ICIs plus conventional therapy resulted in a significantly greater risk of hypothyroidism than treatment with one ICI plus conventional therapy. Among the ICI monotherapies, nivolumab, pembrolizumab, camrelizumab, and durvalumab were associated with significantly greater incidence of hypothyroidism than conventional therapy, whereas ipilimumab, avelumab, and atezolizumab were not associated with elevated risk of hypothyroidism. Nivolumab and pembrolizumab were associated with significantly greater risk of hypothyroidism than ipilimumab, whereas no significant difference was observed in the incidence of hypothyroidism with treatment with other ICI monotherapy strategies. SUCRA analysis indicated that ipilimumab had the best safety ranking for hypothyroidism (probability = 78.6%) among treatment regimens involving ICIs, and was followed by avelumab (77.3%), one ICI in combination with traditional treatment (67.7%), two ICIs (58.4%), atezolizumab (48.8%), nivolumab (38.7%), pembrolizumab (34.6%), cemiplimab (27.6%), durvalumab (12.2%), and dual ICIs in combination with conventional therapy (9.1%) ( Figure 3b ).

            Figure 3 |

            Comparative analysis of hypothyroidism in a consistency model-based network meta-analysis (NMA) (A) league table and (B) probability ranking chart. Panel A presents a league table delineating the pooled odds ratios (OR) and corresponding 95% confidence intervals (CI) for drug-induced immune-related hypothyroidism across different treatment regimens. Statistically significant outcomes are highlighted in bold. Panel B depicts probability ranking curves, which quantify the likelihood of each treatment achieving a specified rank in terms of hypothyroidism risk reduction, ranging from lowest to highest. Abbreviations: ICI = immune checkpoint inhibitor.

            3.3.3.2 Hyperthyroidism

            Similarly to the results for hypothyroidism, ICI combination therapies, compared with conventional therapy, were associated with varying degrees of hyperthyroidism risk ( Figure 4a ). The risk of hyperthyroidism was higher with dual ICIs plus conventional therapy than with one ICI plus conventional therapy. Monotherapy a PD-1 inhibitor (nivolumab or pembrolizumab) or CTLA-4 inhibitor (ipilimumab) was associated with significantly greater risk of hyperthyroidism than conventional therapy, whereas treatment with a PD-L1 inhibitor (atezolizumab) was not associated with increased risk. Moreover, nivolumab and pembrolizumab were associated with a significantly greater risk of hyperthyroidism than ipilimumab. The safety ranking for hyperthyroidism among treatment regimens involving ICIs indicated that PD1/PD-L1 inhibitor plus conventional therapy had the highest probability (78.6%), followed by pembrolizumab (67.7%), nivolumab (73.3%), two ICIs (60.4%), ipilimumab (37.3%), and two ICIs plus conventional therapy (5.9%) ( Figure 4b ).

            Figure 4 |

            Comparative analysis of hyperthyroidism in a consistency model-based network meta-analysis (NMA) (A) league table and (B) probability ranking chart. Panel A presents a league table delineating the pooled odds ratios (OR) and corresponding 95% confidence intervals (CI) for drug-induced immune-related hypothyroidism across different treatment regimens. Statistically significant outcomes are highlighted in bold. Panel B depicts probability ranking curves, which quantify the likelihood of each treatment achieving a specified rank in terms of hypothyroidism risk reduction, ranging from lowest to highest. Abbreviations: ICI = immune checkpoint inhibitor.

            3.3.3.3 Hypophysitis

            On the basis of the findings presented in Figure 5a , ipilimumab and pembrolizumab were associated with significantly greater risk of hypophysitis than conventional therapy. Moreover, ipilimumab was associated with a greater risk of hypophysitis than pembrolizumab and nivolumab, or PD1/PD-L1 plus conventional therapy. Furthermore, treatment with two ICIs was associated with a greater risk of hypophysitis than treatment with one ICI plus conventional therapy, as well as ipilimumab, pembrolizumab, or nivolumab monotherapies. In the cumulative probability ranking for hypophysitis ( Figure 5b ), conventional therapy had the best safety ranking (95.6%), and was followed by nivolumab (70.0%), one ICI plus traditional therapy (67.5%), pembrolizumab (55.9%), tislelizumab (33.2%), ipilimumab (24.1%), and dual ICI therapy (3.7%).

            Figure 5 |

            Comparative analysis of hypophysitis in a consistency model-based network meta-analysis (NMA) (A) league table and (B) probability ranking chart. Panel A presents a league table delineating the pooled odds ratios (OR) and corresponding 95% confidence intervals (CI) for drug-induced immune-related hypothyroidism across different treatment regimens. Statistically significant outcomes are highlighted in bold. Panel B depicts probability ranking curves, which quantify the likelihood of each treatment achieving a specified rank in terms of hypothyroidism risk reduction, ranging from lowest to highest. Abbreviations: ICI = immune checkpoint inhibitor.

            3.3.3.4 Adrenal insufficiency

            The use of combination therapy with two ICIs, or two ICIs plus conventional therapy, was associated with greater risk of adrenal insufficiency than conventional therapy ( Figure 6a ). Ipilimumab and pembrolizumab were also associated with significantly greater risk of adrenal insufficiency than traditional therapy, whereas nivolumab was not associated with a significantly elevated risk. Moreover, no significant difference was observed in the incidence of adrenal insufficiency among ICI monotherapies. The cumulative probability ranking of adrenal insufficiency, ordered from highest to lowest ( Figure 6b ), indicated that conventional therapy had the best safety ranking, at 97.5%, and was followed by one ICI in combination with traditional therapy (74.0%), pembrolizumab (58.3%), nivolumab (54.5%), two ICIs in combination (32.2%), ipilimumab (17.9%), and two ICIs in combination with traditional therapy (15.5%).

            Figure 6 |

            Comparative analysis of adrenal insufficiency in a consistency model-based network meta-analysis (NMA) (A) league table and (B) probability ranking chart. Panel A presents a league table delineating the pooled odds ratios (OR) and corresponding 95% confidence intervals (CI) for drug-induced immune-related hypothyroidism across different treatment regimens. Statistically significant outcomes are highlighted in bold. Panel B depicts probability ranking curves, which quantify the likelihood of each treatment achieving a specified rank in terms of hypothyroidism risk reduction, ranging from lowest to highest. Abbreviations: ICI = immune checkpoint inhibitor.

            3.3.3.5 Thyroiditis

            Treatment with one ICI plus conventional therapy, or two ICIs, was associated with significantly greater risk of thyroiditis than conventional therapy ( Figure 7a ). Additionally, pembrolizumab was associated with greater risk of thyroiditis than conventional therapy. SUCRA analysis also indicated that treatment with two ICIs had the highest ranking regarding thyroiditis (8.4%) ( Figure 7b ).

            Figure 7 |

            Comparative analysis of thyroiditis in a consistency model-based network meta-analysis (NMA) (A) league table and (B) probability ranking chart. Panel A presents a league table delineating the pooled odds ratios (OR) and corresponding 95% confidence intervals (CI) for drug-induced immune-related hypothyroidism across different treatment regimens. Statistically significant outcomes are highlighted in bold. Panel B depicts probability ranking curves, which quantify the likelihood of each treatment achieving a specified rank in terms of hypothyroidism risk reduction, ranging from lowest to highest. Abbreviations: ICI = immune checkpoint inhibitor.

            3.3.4 Subgroup analysis
            3.3.4.1 Melanoma

            We conducted a subgroup analysis of endocrine toxicity, including hypothyroidism, hyperthyroidism, and hypophysitis, in patients with melanoma treated with ICIs ( Figures S7–S9 ).

            3.3.4.1.1 Hypothyroidism

            According to the results shown in Figure S7A , treatments with one ICI plus conventional therapy, or with monotherapies comprising nivolumab or pembrolizumab, were associated with greater incidence of hypothyroidism than conventional therapy alone. Moreover, a combination of two ICIs or pembrolizumab alone was associated with a significantly greater risk than treatment with ipilimumab alone. Other treatment comparisons did not reveal any notable differences. According to SUCRA analysis ( Figure S7B ), conventional therapy (95.7%) was safest, and was followed by ipilimumab (81.0%), two ICIs (44.6%), one ICI plus conventional therapy (35.8%), nivolumab (31.8%), and pembrolizumab (11.1%).

            3.3.4.1.2 Hyperthyroidism

            Regarding hyperthyroidism, no significant differences were observed in the incidence of hypothyroidism among strategies ( Figure S8A ). The safety ranking for hyperthyroidism among treatment regimens involving ICIs indicated that ipilimumab (88.6%) had the highest probability, and was followed by pembrolizumab (49.2%), two ICIs (42.1%), and nivolumab (20.1%) ( Figure S8B ).

            3.3.4.1.3 Hypophysitis

            Regarding hypophysitis, both ipilimumab and dual ICI therapies were associated with significantly greater risk than one ICI plus conventional therapy, nivolumab, and pembrolizumab ( Figure S9A ). Notably, the risk with dual ICI therapy was significantly higher than that with ipilimumab alone. No significant differences emerged in other treatment comparisons. The cumulative probability curve for hypophysitis, ranked from highest to lowest risk, was as follows: one ICI plus conventional therapy (83.7%), nivolumab (77.5%), pembrolizumab (62.6%), ipilimumab (25.8%), and two ICIs (0.4%) ( Figure S9B ).

            3.3.4.2 NSCLC

            A subgroup analysis was also conducted for patients with NSCLC and four endocrine toxicities: hypothyroidism, hyperthyroidism, hypophysitis, and thyroiditis ( Figures S10–S13 ).

            3.3.4.2.1 Hypothyroidism

            One ICI plus conventional therapy, pembrolizumab, tremelimumab, two ICIs plus conventional therapy, nivolumab, and dual ICI therapy were all associated with greater incidence of hypothyroidism than conventional therapy ( Figure S10A ). Notably, pembrolizumab, two ICIs plus conventional therapy, and two ICIs were associated with a significantly greater risk of hypothyroidism than one ICI plus conventional therapy. No significant differences were observed in other treatment comparisons. The cumulative probability curve area for hypothyroidism, ranked from lowest to highest risk, was as follows: conventional therapy (99.5%), one ICI plus conventional therapy (78.1%), atezolizumab (70.6%), pembrolizumab (54.2%), tremelimumab (38.7%), two ICIs plus conventional therapy (23.9%), nivolumab (21.3%), and dual ICI therapy (13.6%) ( Figure S10B ).

            3.3.4.2.2 Hyperthyroidism

            In the context of hyperthyroidism, pembrolizumab was associated with significantly greater risk than treatment with one ICI plus conventional therapy or conventional therapy alone ( Figure S11A ). Other treatment comparisons did not show significant differences. The cumulative probability curve for hyperthyroidism, from highest to lowest risk, was as follows: conventional therapy (87.6%), atezolizumab (60.1%), one ICI plus conventional therapy (46.9%), and pembrolizumab (5.4%) ( Figure S11B ).

            3.3.4.2.3 Hypophysitis

            Our analysis revealed no significant differences in hypophysitis risk across various treatment modalities ( Figure S12A ). The cumulative probability curve for hypophysitis, from highest to lowest risk, was as follows: conventional therapy (91.9%), pembrolizumab (38.2%), and one ICI plus conventional therapy (19.9%) ( Figure S12B ).

            3.3.4.2.4 Thyroiditis

            On the basis of the findings presented in Figure S13A , pembrolizumab was associated with notably greater risk than conventional therapy. No other treatment comparisons showed significant differences. SUCRA analysis indicated that conventional therapy (93.0%) was the safest option regarding thyroiditis among the ICI regimens, followed by one ICI plus conventional therapy (42.1%) and pembrolizumab (14.9%) ( Figure S13B ).

            3.3.5 Sensitivity analysis

            To assess the stability of the NMA results, we conducted a sensitivity analysis by systematically excluding each study from the safety data set. Although most of the sensitivity analysis data aligned with the broader trend, several exceptions, such as those from the E1609 and CheckMate 238 studies, deviated from the conventional range. These deviations might have arisen from specific sample characteristics, stringent selection criteria, or unique methodological approaches. For instance, the E1609 study used a two-step hierarchical approach for assessing ipilimumab’s efficacy and safety at various dosages in patients with melanoma. These specialized trial designs might have introduced selection bias if their initial screening phases were not randomized, thus potentially affecting the generalizability of the findings. Nonetheless, our overall conclusions indicated resilience against individual study variances, and a consistent overall trend was maintained and was unaffected by the exceptions ( Figures S14–S18 ).

            3.3.6 Bias detection

            Regarding bias detection, the funnel plot CI for six studies on hypothyroidism exceeded the range; one study on hyperthyroidism also exceeded the range, potentially because of heterogeneity among studies. In contrast, the remaining studies showed basic symmetry, thereby suggesting a possible small sample size effect or relatively low risk of publication bias, as depicted in Figure S19 .

            4. DISCUSSION

            The advent of ICIs has revolutionized cancer therapeutics and ushered in a new era of effective treatments that mobilize the immune system to target malignancies. Although this progress is encouraging, the risks of irAEs, specifically endocrine toxicity, remain an underexplored challenge that merits rigorous investigation [7375]. Endocrine toxicity, one of the most common irAE types, requires attention throughout the entire course of cancer treatment [76]. Evidence-based synthesis and evaluation of endocrine toxicity associated with various ICI treatment strategies via NMA can provide clinicians and patients with a more informed perspective regarding the risks and benefits of various ICI treatment options.

            Although prior meta-analyses have reported elevated risks of endocrine dysfunction associated with ICIs compared with chemotherapy or placebos, our study is unprecedented in its comprehensive comparison of specific ICI regimens. Unlike previous studies with a narrower focus, our NMA accommodates the newest ICI therapies, including camrelizumab, the first PD-1 inhibitor approved in China for liver cancer, and combination therapies, such as durvalumab plus tremelimumab. This broad analysis enables clinicians to gauge endocrine toxicity risks across multiple ICI therapies, thus providing an invaluable tool for individualized treatment planning.

            Our study, performing NMA on 55 RCTs involving 32,522 patients, provides a detailed exploration of endocrine toxicity across a spectrum of ICI treatments. Our findings reaffirm the heightened risk of endocrine toxicity in patients receiving ICI therapy, with particular emphasis on the unique toxicity profiles of different ICIs and their combinations. First, the study demonstrated that dual ICI combinations generally pose greater risks than monotherapies, probably because of synergistic immune activation and longer treatment durations [77, 78]. This finding has immediate ramifications for treatment protocols, by indicating the need for vigilant monitoring and preemptive interventions to mitigate toxicity. Second, PD1/PD-L1 inhibitors are more likely than CTLA-4 inhibitors to induce thyroid-related adverse events, such as hypothyroidism and hyperthyroidism. This finding might be due to the high expression of PD-L1 in the follicular cells of the thyroid gland; this expression is positively correlated with the severity of autoimmune thyroid diseases [79, 80]. In contrast, CTLA-4 inhibitors were associated with hypophysitis, a finding indicating a need for specialized monitoring and therapeutic strategies for patients receiving these drugs. Previous studies have shown that CTLA-4 protein is expressed in the pituitary gland, which plays a key role in regulating hormone secretion [81, 82]. Therefore, CTLA-4 inhibitors may interfere with the normal function of the pituitary gland and lead to a decrease in, or cessation of, anterior pituitary hormone secretion [8385]. In addition, CTLA-4 inhibitors may cause expansion and activation of T cells, and consequently exacerbate the body’s autoimmune response and aggravate the severity of hypophysitis. However, tremelimumab, another CTLA-4 inhibitor, has shown a better safety profile and was not found to significantly increase the risk of hypophysitis. Notably, the studies included in the analysis of tremelimumab are relatively limited, and further research is required to confirm this conclusion. These granular insights may facilitate more precise tailoring of treatments, thereby enabling clinicians to make educated therapeutic choices while considering individual patient vulnerabilities.

            Notably, nivolumab had a high safety profile, comparable to that of conventional therapy, in terms of adrenal insufficiency. In contrast, pembrolizumab, ipilimumab, two ICIs, or two ICIs plus conventional therapy had a significantly greater risk of inducing adrenal dysfunction. Moreover, pembrolizumab and PD1/PD-L1 plus conventional therapy or two ICIs increased the risk of thyroiditis, although to varying degrees.

            In our comprehensive analysis of endocrine toxicity associated with ICIs, we extensively reviewed and contrasted the findings from a broad array of previous studies with our own results. Initially, we examined articles addressing the overall safety of ICIs, particularly those detailing outcomes associated with endocrine functions. For instance, an NMA on the overall safety of ICI in cancer treatment has demonstrated that nivolumab is associated with a greater risk of hypothyroidism than ipilimumab, a finding consistent with our results [20]. Furthermore, another study conducting an NMA on the overall safety of ICI in solid tumors has highlighted that durvalumab combined with conventional therapy presents a greater risk of hyperthyroidism than pembrolizumab with conventional therapy [86]. Our more detailed study specifically focused on comparing durvalumab and pembrolizumab as monotherapies, and identified no significant difference in the risk of hyperthyroidism between these treatments.

            Expanding our focus, we analyzed research on ICI-related adverse endocrine reactions. We discovered a lack of network meta-analyses directly targeting ICI-induced endocrine adverse reactions, thus emphasizing the innovative and crucial nature of our work. Our comparison of our network meta-analysis with several existing meta-analyses revealed both consistencies and differences. PD-1/PD-L1 inhibitors were consistently implicated in a higher incidence of thyroid-related adverse events, such as hypothyroidism and hyperthyroidism, than CTLA-4 inhibitors [20, 87]. This trend is corroborated by multiple studies reporting a significant risk of hypophysitis associated with ipilimumab [87]. Nonetheless, our study’s conclusions diverge from the findings in some previous research. For instance, whereas some studies have indicated a greater risk of hyperthyroidism with PD-1 inhibitors than PD-L1 inhibitors, our analysis did not indicate this distinction [87]. Given the expanded number of studies included in our research, our findings must be juxtaposed with those of prior research, to identify and discuss the nuances and broader implications of these similarities and differences.

            However, several study limitations must be acknowledged. First, our study focused on English-language publications and did not analyze influential factors such as sex, age, and geographical distribution. Second, the scarcity of studies specifically investigating endocrine adverse events induced by durvalumab or tremelimumab necessitates further research with larger sample sizes to substantiate the findings regarding the incidence of these events after treatment with these drugs. Third, our research demonstrated low overall inconsistency yet relatively high heterogeneity among studies. This finding might have been due to the biological diversity of cancer types, variations in patient demographics, the complexity of treatment regimens, and differing statistical methods. Future efforts will be aimed at exploring the sources of this heterogeneity, to minimize heterogeneity and bolster the rigor of our findings.

            Additionally, most included studies were clinical trials for drug registration, involving patients selected based on strict criteria. This selection might limit the applicability of our results to a broader patient spectrum, including older individuals and those with multiple comorbidities. These inherent limitations underscore the need for further more inclusive research.

            Despite these constraints, our study notably carefully compared endocrine toxicity risks across various ICI regimens, thus adding depth to the growing body of ICI research. Our findings equip clinicians with a sophisticated tool for risk assessment and management, thereby augmenting the safety and efficacy of ICI treatments. Future research should build on this groundwork, integrating real-world data and focusing on underrepresented populations to foster a comprehensive understanding of ICI-related endocrine toxicity.

            5. CONCLUSION

            In conclusion, ICIs have been associated with an elevated risk of endocrine toxicity—a risk further amplified with the use of dual ICI regimens. Specifically, CTLA-4 inhibitors are notably associated with hypophysitis, whereas PD-1/PD-L1 inhibitors are more commonly associated with thyroid-related conditions such as hyperthyroidism, hypothyroidism, and thyroiditis. Interestingly, nivolumab has a safety profile similar to that of conventional therapies in terms of adrenal dysfunction, in contrast to the heightened risks associated with other ICI treatments. In the context of melanoma, the safety profile of ICIs generally aligns with broader study findings. Pembrolizumab tends to induce hypothyroidism more frequently than ipilimumab, whereas ipilimumab is associated with a greater risk of hypophysitis than PD-1/PD-L1 inhibitors, such as pembrolizumab and nivolumab. In NSCLC, pembrolizumab is associated with a relatively lower safety profile including elevated risks of hypothyroidism, hyperthyroidism, and thyroiditis. Our study provides crucial evidence-based insights aimed at refining the risk-benefit calculus and contributing to the development of safer and more efficacious ICI therapies in clinical settings.

            Supplementary Material

            Supplementary Material can be downloaded here

            REFERENCES

            1. Yang H, Yao Z, Zhou X, Zhang W, Zhang X, Zhang F. Immune-Related Adverse Events of Checkpoint Inhibitors: Insights Into Immunological Dysregulation. Clinical Immunology. 2020. Vol. 213:108377

            2. Quandt Z, Young A, Perdigoto AL, Herold KC, Anderson MS. Autoimmune Endocrinopathies: An Emerging Complication of Immune Checkpoint Inhibitors. Annual Review of Medicine. 2021. Vol. 72:313–330

            3. Bagchi S, Yuan R, Engleman EG. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annual Review of Pathology. 2021. Vol. 16:223–249

            4. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al.. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. The New England Journal of Medicine. 2014. Vol. 371:2189–2199

            5. Tang Q, Chen Y, Li X, Long S, Shi Y, Yu Y, et al.. The Role of PD-1/PD-L1 and Application of Immune-Checkpoint Inhibitors in Human Cancers. Frontiers in Immunology. 2022. Vol. 13:964442

            6. Marei HE, Hasan A, Pozzoli G, Cenciarelli C. Cancer Immunotherapy with Immune Checkpoint Inhibitors (ICIs): Potential, Mechanisms of Resistance, and Strategies for Reinvigorating T Cell Responsiveness when Resistance is Acquired. Cancer Cell International. 2023. Vol. 23:64

            7. Kao HF, Lou PJ. Immune Checkpoint Inhibitors for Head and Neck Squamous Cell Carcinoma: Current Landscape and Future Directions. Head & Neck. 2019. Vol. 41 Suppl 1:4–18

            8. Fiorentino V, Tralongo P, Larocca LM, Pizzimenti C, Martini M, Pierconti F. First-Line ICIs in Renal Cell Carcinoma. Human Vaccines & Immunotherapeutics. 2023. Vol. 19:2225386

            9. Wright JJ, Powers AC, Johnson DB. Endocrine Toxicities of Immune Checkpoint Inhibitors. Nature Reviews Endocrinology. 2021. Vol. 17:389–399

            10. Tison A, Garaud S, Chiche L, Cornec D, Kostine M. Immune-Checkpoint Inhibitor Use in Patients with Cancer and Pre-Existing Autoimmune Diseases. Nature Reviews Endocrinology. 2022. Vol. 18:641–656

            11. Vafaei S, Zekiy AO, Khanamir RA, Zaman BA, Ghayourvahdat A, Azimizonuzi H, et al.. Combination Therapy with Immune Checkpoint Inhibitors (ICIs); A New Frontier. Cancer Cell International. 2022. Vol. 22:2

            12. Wang M, Liu Y, Cheng Y, Wei Y, Wei X. Immune Checkpoint Blockade and its Combination Therapy with Small-Molecule Inhibitors for Cancer Treatment. Biochimica et Biophysica Acta - Reviews on Cancer. 2019. Vol. 1871:199–224

            13. Kgatle MM, Boshomane TMG, Lawal IO, Mokoala KMG, Mokgoro NP, Lourens N, et al.. Immune Checkpoints, Inhibitors and Radionuclides in Prostate Cancer: Promising Combinatorial Therapy Approach. International Journal of Molecular Sciences. 2021. Vol. 22:4109

            14. Grasselly C, Denis M, Bourguignon A, Talhi N, Mathe D, Tourette A, et al.. The Antitumor Activity of Combinations of Cytotoxic Chemotherapy and Immune Checkpoint Inhibitors is Model-Dependent. Frontiers in Immunology. 2018. Vol. 9:2100

            15. Choueiri TK, Powles T, Burotto M, Escudier B, Bourlon MT, Zurawski B, et al.. Nivolumab plus Cabozantinib versus Sunitinib for Advanced Renal-Cell Carcinoma. The New England Journal of Medicine. 2021. Vol. 384:829–841

            16. Motzer R, Alekseev B, Rha SY, Porta C, Eto M, Powles T, et al.. Lenvatinib plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. The New England Journal of Medicine. 2021. Vol. 384:1289–1300

            17. Barnabei A, Senes P, Scoppola A, Chiefari A, Iannantuono GM, Appetecchia M, et al.. Endocrine Toxicities of Antineoplastic Therapy: The Adrenal Topic. Cancers (Basel). 2022. Vol. 14:593

            18. Moc C. Endocrinopathies from Checkpoint Inhibitors: Incidence, Outcomes, and Management. Cleveland Clinic Journal of Medicine. 2023. Vol. 90:307

            19. Xie J, Zhang Z, Zhang S, Lv Y, Mao Y, Liu R, et al.. Odds Ratio of Programmed Cell Death-1 or Ligand 1 Inhibitor-Related Endocrine Dysfunction in Patients with Lung Cancer: A Systematic Review and Meta-Analysis. Medicine (Baltimore). 2019. Vol. 98:e18310

            20. Xu C, Chen YP, Du XJ, Liu JQ, Huang CL, Chen L, et al.. Comparative Safety of Immune Checkpoint Inhibitors in Cancer: Systematic Review and Network Meta-Analysis. BMJ. 2018. Vol. 363:k4226

            21. Yang Y, Liu J, Yang K, Ma Y, Fu S, Tang X, et al.. Endocrine Adverse Events Caused by Different Types and Different Doses of Immune Checkpoint Inhibitors in the Treatment of Solid Tumors: A Meta-Analysis and Systematic Review. The Journal of Clinical Pharmacology. 2021. Vol. 61:282–297

            22. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al.. KEYNOTE-006 Investigators: Pembrolizumab versus Ipilimumab in Advanced Melanoma. The New England Journal of Medicine. 2015. Vol. 372:2521–2532

            23. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al.. OAK Study Group: Atezolizumab versus Docetaxel in Patients with Previously Treated Non-Small-Cell Lung Cancer (OAK): A Phase 3, Open-Label, Multicentre Randomised Controlled Trial. Lancet. 2017. Vol. 389:255–265

            24. Larkin J, Minor D, D’Angelo S, Neyns B, Smylie M, Miller WH Jr, et al.. Overall Survival in Patients with Advanced Melanoma who Received Nivolumab Versus Investigator’s Choice Chemotherapy in CheckMate 037: A Randomized, Controlled, Open-Label Phase III Trial. Journal of Clinical Oncology. 2018. Vol. 36:383–390

            25. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al.. CheckMate 025 Investigators: Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. The New England Journal of Medicine. 2015. Vol. 373:1803–1813

            26. Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee JL, Fong L, et al.. KEYNOTE-045 Investigators: Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. The New England Journal of Medicine. 2017. Vol. 376:1015–1026

            27. Bang YJ, Cho JY, Kim YH, Kim JW, Di Bartolomeo M, Ajani JA, et al.. Efficacy of Sequential Ipilimumab Monotherapy versus Best Supportive Care for Unresectable Locally Advanced/Metastatic Gastric or Gastroesophageal Junction Cancer. Clinical Cancer Research. 2017. Vol. 23:5671–5678

            28. Ascierto PA, Del Vecchio M, Robert C, Mackiewicz A, Chiarion-Sileni V, Arance A, et al.. Ipilimumab 10 mg/kg versus Ipilimumab 3 mg/kg in Patients with Unresectable or Metastatic Melanoma: A Randomised, Double-Blind, Multicentre, Phase 3 Trial. The Lancet Oncology. 2017. Vol. 18:611–622

            29. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al.. KEYNOTE-024 Investigators: Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. The New England Journal of Medicine. 2016. Vol. 375:1823–1833

            30. Langer CJ, Gadgeel SM, Borghaei H, Papadimitrakopoulou VA, Patnaik A, Powell SF, et al.. KEYNOTE-021 investigators: Carboplatin and Pemetrexed with or Without Pembrolizumab for Advanced, Non-Squamous Non-Small-Cell Lung Cancer: A Randomised, Phase 2 Cohort of the Open-Label KEYNOTE-021 Study. The Lancet Oncology. 2016. Vol. 17:1497–1508

            31. Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, et al.. Nivolumab and Ipilimumab versus Ipilimumab in Untreated Melanoma. The New England Journal of Medicine. 2015. Vol. 372:2006–2017

            32. Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, et al.. Pembrolizumab versus Docetaxel for Previously Treated, PD-L1-Positive, Advanced Non-Small-Cell Lung Cancer (KEYNOTE-010): A Randomised Controlled Trial. Lancet. 2016. Vol. 387:1540–1550

            33. Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, et al.. POPLAR Study Group: Atezolizumab versus Docetaxel for Patients with Previously Treated Non-Small-Cell Lung Cancer (POPLAR): A Multicentre, Open-Label, Phase 2 Randomised Controlled Trial. Lancet. 2016. Vol. 387:1837–1846

            34. Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, et al.. Pembrolizumab versus Investigator-Choice Chemotherapy for Ipilimumab-Refractory Melanoma (KEYNOTE-002): A Randomised, Controlled, Phase 2 Trial. The Lancet Oncology. 2015. Vol. 16:908–918

            35. Ribas A, Kefford R, Marshall MA, Punt CJ, Haanen JB, Marmol M, et al.. Phase III Randomized Clinical Trial Comparing Tremelimumab with Standard-of-Care Chemotherapy in Patients with Advanced Melanoma. Journal of Clinical Oncology. 2013. Vol. 31:616–622

            36. Powles T, Park SH, Voog E, Caserta C, Valderrama BP, Gurney H, et al.. Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma. The New England Journal of Medicine. 2020. Vol. 383:1218–1230

            37. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, et al.. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. The New England Journal of Medicine. 2019. Vol. 380:1103–1115

            38. Hodi FS, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Cowey CL, et al.. Nivolumab plus Ipilimumab or Nivolumab alone versus Ipilimumab alone in Advanced Melanoma (CheckMate 067): 4-year Outcomes of a Multicentre, Randomised, Phase 3 Trial. The Lancet Oncology. 2018. Vol. 19:1480–1492

            39. Lebbé C, Meyer N, Mortier L, Marquez-Rodas I, Robert C, Rutkowski P, et al.. Evaluation of Two Dosing Regimens for Nivolumab in Combination with Ipilimumab in Patients with Advanced Melanoma: Results From the Phase IIIb/IV CheckMate 511 Trial. Journal of Clinical Oncology. 2019. Vol. 37:867–875

            40. Zamarin D, Burger RA, Sill MW, Powell DJ Jr, Lankes HA, Feldman MD, et al.. Randomized Phase II Trial of Nivolumab Versus Nivolumab and Ipilimumab for Recurrent or Persistent Ovarian Cancer: An NRG Oncology Study. Journal of Clinical Oncology. 2020. Vol. 38:1814–1823. Journal of Clinical Oncology

            41. Ferris RL, Haddad R, Even C, Tahara M, Dvorkin M, Ciuleanu TE, et al.. Durvalumab with or without Tremelimumab in Patients with Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: EAGLE, a Randomized, Open-Label Phase III Study. Annals of Oncology. 2020. Vol. 31:942–950

            42. Long GV, Dummer R, Hamid O, Gajewski TF, Caglevic C, Dalle S, et al.. Epacadostat plus Pembrolizumab versus Placebo plus Pembrolizumab in Patients with Unresectable or Metastatic Melanoma (ECHO-301/KEYNOTE-252): A Phase 3, Randomised, Double-Blind Study. The Lancet Oncology. 2019. Vol. 20:1083–1097

            43. Rozeman EA, Menzies AM, van Akkooi ACJ, Adhikari C, Bierman C, van de Wiel BA, et al.. Identification of the Optimal Combination Dosing Schedule of Neoadjuvant Ipilimumab plus Nivolumab in Macroscopic Stage III melanoma (OpACIN-neo): A Multicentre, Phase 2, Randomised, Controlled Trial. The Lancet Oncology. 2019. Vol. 20:948–960

            44. Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J, et al.. KEYNOTE-407 Investigators: Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. The New England Journal of Medicine. 2018. Vol. 379:2040–2051

            45. Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al.. KEYNOTE-042 Investigators: Pembrolizumab versus Chemotherapy for Previously Untreated, PD-L1-Expressing, Locally Advanced or Metastatic Non-Small-Cell Lung Cancer (KEYNOTE-042): A Randomised, Open-Label, Controlled, Phase 3 Trial. Lancet. 2019. Vol. 393:1819–1830

            46. Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M, de Castro G Jr, et al.. KEYNOTE-048 Investigators: Pembrolizumab Alone or with Chemotherapy versus Cetuximab with Chemotherapy for Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck (KEYNOTE-048): A Randomised, Open-Label, Phase 3 Study. Lancet. 2019. Vol. 394:1915–1928

            47. Gadgeel S, Rodríguez-Abreu D, Speranza G, Esteban E, Felip E, Dómine M, et al.. Updated Analysis From KEYNOTE-189: Pembrolizumab or Placebo Plus Pemetrexed and Platinum for Previously Untreated Metastatic Nonsquamous Non-Small-Cell Lung Cancer. Journal of Clinical Oncology. 2020. Vol. 38:1505–1517

            48. Schmid P, Cortes J, Pusztai L, McArthur H, Kümmel S, Bergh J, et al.. KEYNOTE-522 Investigators: Pembrolizumab for Early Triple-Negative Breast Cancer. The New England Journal of Medicine. 2020. Vol. 382:810–821

            49. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al.. Updated Analysis of KEYNOTE-024: Pembrolizumab Versus Platinum-Based Chemotherapy for Advanced Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score of 50% or Greater. Journal of Clinical Oncology. 2019. Vol. 37:537–546

            50. Schmid P, Rugo HS, Adams S, Schneeweiss A, Barrios CH, Iwata H, et al.. IMpassion130 Investigators: Atezolizumab plus Nab-Paclitaxel as First-Line Treatment for Unresectable, Locally Advanced or Metastatic Triple-Negative Breast Cancer (IMpassion130): Updated Efficacy Results from a Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. The Lancet Oncology. 2020. Vol. 21:44–59

            51. André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, et al.. KEYNOTE-177 Investigators: Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. The New England Journal of Medicine. 2020. Vol. 383:2207–2218

            52. Motzer RJ, Rini BI, McDermott DF, Arén Frontera O, Hammers HJ, Carducci MA, et al.. CheckMate 214 Investigators: Nivolumab Plus Ipilimumab Versus Sunitinib in First-Line Treatment for Advanced Renal Cell Carcinoma: Extended Follow-Up of Efficacy and Safety Results From a Randomised, Controlled, Phase 3 Trial. The Lancet Oncology. 2019. Vol. 20:1370–1385

            53. Cohen EEW, Soulières D, Le Tourneau C, Dinis J, Licitra L, Ahn MJ, et al.. KEYNOTE-040 Investigators: Pembrolizumab Versus Methotrexate, Docetaxel, or Cetuximab for Recurrent or Metastatic Head-and-Neck Squamous Cell Carcinoma (KEYNOTE-040): A Randomised, Open-Label, Phase 3 Study. Lancet. 2019. Vol. 393:156–167

            54. Rini BI, Powles T, Atkins MB, Escudier B, McDermott DF, Suarez C, et al.. IMmotion151 Study Group: Atezolizumab Plus Bevacizumab Versus Sunitinib in Patients with Previously Untreated Metastatic Renal Cell Carcinoma (IMmotion151): A Multicentre, Open-Label, Phase 3, Randomised Controlled Trial. Lancet. 2019. Vol. 393:2404–2415

            55. Fradet Y, Bellmunt J, Vaughn DJ, Lee JL, Fong L, Vogelzang NJ, et al.. Randomized Phase III KEYNOTE-045 Trial of Pembrolizumab Versus Paclitaxel, Docetaxel, or Vinflunine in Recurrent Advanced Urothelial Cancer: Results of >2 Years of Follow-Up. Annals of Oncology. 2019. Vol. 30:970–976

            56. Shitara K, Van Cutsem E, Bang YJ, Fuchs C, Wyrwicz L, Lee KW, et al.. Efficacy and Safety of Pembrolizumab or Pembrolizumab Plus Chemotherapy vs Chemotherapy Alone for Patients With First-line, Advanced Gastric Cancer: The KEYNOTE-062 Phase 3 Randomized Clinical Trial. JAMA Oncology. 2020. Vol. 6:1571–1580

            57. Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, et al.. Atezolizumab, Vemurafenib, and Cobimetinib as First-Line Treatment for Unresectable Advanced BRAFV600 Mutation-Positive Melanoma (IMspire150): Primary Analysis of the Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet. 2020. Vol. 395:1835–1844

            58. Mittendorf EA, Zhang H, Barrios CH, Saji S, Jung KH, Hegg R, et al.. Neoadjuvant Atezolizumab in Combination with Sequential nab-Paclitaxel and Anthracycline-Based Chemotherapy Versus Placebo and Chemotherapy in Patients with Early-Stage Triple-Negative Breast Cancer (IMpassion031): A Randomised, Double-Blind, Phase 3 Trial. Lancet. 2020. Vol. 396:1090–1100

            59. Reck M, Schenker M, Lee KH, Provencio M, Nishio M, Lesniewski-Kmak K, Sangha R, et al.. Nivolumab Plus Ipilimumab Versus Chemotherapy as First-Line Treatment in Advanced Non-Small-Cell Lung Cancer with High Tumour Mutational Burden: Patient-Reported Outcomes Results from the Randomised, Open-Label, Phase III CheckMate 227 Trial. European Journal of Cancer. 2019. Vol. 116:137–147

            60. Paz-Ares L, Ciuleanu TE, Cobo M, Schenker M, Zurawski B, Menezes J, et al.. First-Line Nivolumab Plus Ipilimumab Combined with Two Cycles of Chemotherapy in Patients with Non-Small-Cell Lung Cancer (CheckMate 9LA): An International, Randomised, Open-Label, Phase 3 trial. The Lancet Oncology. 2021. Vol. 22:198–211

            61. Powles T, Plimack ER, Soulières D, Waddell T, Stus V, Gafanov R, et al.. Pembrolizumab Plus Axitinib Versus Sunitinib Monotherapy as First-Line Treatment of Advanced Renal Cell Carcinoma (KEYNOTE-426): Extended Follow-Up from a Randomised, Open-Label, Phase 3 Trial. The Lancet Oncology. 2020. Vol. 21:1563–1573

            62. Rudin CM, Awad MM, Navarro A, Gottfried M, Peters S, Csőszi, et al.. KEYNOTE-604 Investigators: Pembrolizumab or Placebo Plus Etoposide and Platinum as First-Line Therapy for Extensive-Stage Small-Cell Lung Cancer: Randomized, Double-Blind, Phase III KEYNOTE-604 Study. Journal of Clinical Oncology. 2020. Vol. 38:2369–2379

            63. Ascierto PA, Long GV, Robert C, Brady B, Dutriaux C, Di Giacomo AM, et al.. Survival Outcomes in Patients With Previously Untreated BRAF Wild-Type Advanced Melanoma Treated With Nivolumab Therapy: Three-Year Follow-Up of a Randomized Phase 3 Trial. JAMA Oncology. 2019. Vol. 5:187–194

            64. Nanda R, Liu MC, Yau C, Shatsky R, Pusztai L, Wallace A, et al.. Effect of Pembrolizumab Plus Neoadjuvant Chemotherapy on Pathologic Complete Response in Women With Early-Stage Breast Cancer: An Analysis of the Ongoing Phase 2 Adaptively Randomized I-SPY2 Trial. JAMA Oncology. 2020. Vol. 6:676–684

            65. Ascierto PA, Del Vecchio M, Mandalá M, Gogas H, Arance AM, Dalle S, et al.. Adjuvant Nivolumab Versus Ipilimumab in Resected Stage IIIB-C and Stage IV Melanoma (CheckMate 238): 4-Year Results From A Multicentre, Double-Blind, Randomised, Controlled, Phase 3 Trial. The Lancet Oncology. 2020. Vol. 21:1465–1477

            66. Bang YJ, Ruiz EY, Van Cutsem E, Lee KW, Wyrwicz L, Schenker M, et al.. Phase III, Randomised Trial of Avelumab Versus Physician’s Choice of Chemotherapy as Third-Line Treatment of Patients with Advanced Gastric or Gastro-Oesophageal Junction Cancer: Primary Analysis of JAVELIN Gastric 300. Annals of Oncology. 2018. Vol. 29:2052–2060

            67. Goldman JW, Dvorkin M, Chen Y, Reinmuth N, Hotta K, Trukhin D, et al.. CASPIAN Investigators: Durvalumab, with or without Tremelimumab, Plus Platinum-Etoposide Versus Platinum-Etoposide alone in First-Line Treatment Of Extensive-Stage Small-Cell Lung Cancer (Caspian): Updated Results from a Randomised, Controlled, Open-Label, Phase 3 trial. The Lancet Oncology. 2021. Vol. 22:51–65

            68. Eng C, Kim TW, Bendell J, Argilés G, Tebbutt NC, Di Bartolomeo M, et al.. IMblaze370 Investigators: Atezolizumab with or without Cobimetinib Versus Regorafenib in Previously Treated Metastatic Colorectal Cancer (IMblaze370): A Multicentre, Open-Label, Phase 3, Randomised, Controlled Trial. The Lancet Oncology. 2019. Vol. 20:849–861

            69. Tarhini AA, Lee SJ, Hodi FS, Rao UNM, Cohen GI, Hamid O, et al.. Phase III Study of Adjuvant Ipilimumab (3 or 10 mg/kg) Versus High-Dose Interferon Alfa-2b for Resected High-Risk Melanoma: North American Intergroup E1609. Journal of Clinical Oncology. 2020. Vol. 38:567–575

            70. Nie J, Wang C, Liu Y, Yang Q, Mei Q, Dong L, et al.. Addition of Low-Dose Decitabine to Anti-PD-1 Antibody Camrelizumab in Relapsed/Refractory Classical Hodgkin Lymphoma. Journal of Clinical Oncology. 2019. Vol. 37:1479–1489

            71. Arrieta O, Barrón F, Ramírez-Tirado LA, Zatarain-Barrón ZL, Cardona AF, Díaz-García D, et al.. Efficacy and Safety of Pembrolizumab plus Docetaxel vs Docetaxel Alone in Patients with Previously Treated Advanced Non-Small Cell Lung Cancer: The PROLUNG Phase 2 Randomized Clinical Trial. JAMA Oncology. 2020. Vol. 6:856–864

            72. Emens LA, Esteva FJ, Beresford M, Saura C, De Laurentiis M, Kim SB, et al.. Trastuzumab emtansine plus Atezolizumab versus Trastuzumab Emtansine plus Placebo in Previously Treated, HER2-Positive Advanced Breast Cancer (KATE2): A Phase 2, Multicentre, Randomised, Double-Blind Trial. Lancet Oncology. 2020. Vol. 21:1283–1295

            73. Abdelhafeez AAM, Shohdy KS, Ibrahim W. Safety of Combination Immune Checkpoint Inhibitors Compared to Monotherapy; A Systematic Review and Meta-Analysis. Cancer Investigation. 2020. Vol. 38:150–157

            74. Geng R, Tang H, You T, Xu X, Li S, Li Z, et al.. Peripheral CD8+ CD28+ T Lymphocytes Predict the Efficacy and Safety of PD-1/PD-L1 Inhibitors in Cancer Patients. Frontiers in Immunology. 2023. Vol. 14:1125876

            75. Hussaini S, Chehade R, Boldt RG, Raphael J, Blanchette P, Maleki Vareki S, et al.. Association Between Immune-Related Side Effects and Efficacy and Benefit of Immune Checkpoint Inhibitors - A Systematic Review and Meta-Analysis. Cancer Treatment Reviews. 2021. Vol. 92:102134

            76. Chang L-S, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, Min L. Endocrine Toxicity of Cancer Immunotherapy Targeting Immune Checkpoints. Endocrine Reviews. 2019. Vol. 40:17–65

            77. Xing R, Gao J, Cui Q, Wang Q. Strategies to Improve the Antitumor Effect of Immunotherapy for Hepatocellular Carcinoma. Frontiers in Immunology. 2021. Vol. 12:783236

            78. Zhao X, Gao F, Yang J, Fan H, Xie Q, Jiang K, et al.. Risk of Adverse Events in Cancer Patients Receiving Nivolumab with Ipilimumab: A Meta-analysis. Frontiers in Oncology. 2022. Vol. 12:877434

            79. Alvarez-Sierra D, Marin-Sanchez A, Ruiz-Blazquez P, de Jesús Gil C, Iglesias-Felip C, González Ó, et al.. Analysis of the PD-1/PD-L1 Axis in Human Autoimmune Thyroid Disease: Insights Into Pathogenesis and Clues to Immunotherapy Associated Thyroid Autoimmunity. Journal of Autoimmunity. 2019. Vol. 103:102285

            80. D’Andréa G, Lassalle S, Guevara N, Mograbi B, Hofman P. From Biomarkers to Therapeutic Targets: The Promise of PD-L1 in Thyroid Autoimmunity and Cancer. Theranostics. 2021. Vol. 11:1310

            81. Caturegli P, Di Dalmazi G, Lombardi M, Grosso F, Larman HB, Larman T, et al.. Hypophysitis Secondary to Cytotoxic T-Lymphocyte-Associated Protein 4 Blockade: Insights into Pathogenesis from an Autopsy Series. The American Journal of Pathology. 2016. Vol. 186:3225–3235

            82. Iwama S, De Remigis A, Callahan MK, Slovin SF, Wolchok JD, Caturegli P. Pituitary Expression of CTLA-4 Mediates Hypophysitis Secondary to Administration of CTLA-4 Blocking Antibody. Science Translational Medicine. 2014. Vol. 6:230ra45

            83. Angelousi A, Alexandraki K, Tsoli M, Kaltsas G, Kassi E. Hypophysitis (Including IgG4 and Immunotherapy). Neuroendocrinology. 2020. Vol. 110:822–835

            84. Kroiss M, Schilling B, Deutschbein T. Endocrine Side Effects of Cancer Immunotherapy. Deutsche Medizinische Wochenschrift. 2020. Vol. 145:1736–1741

            85. Siewe N, Friedman A. Optimal Timing of Steroid Initiation in Response to CTLA-4 Antibody in Metastatic Cancer: A Mathematical Model. PLos One. 2022. Vol. 17:e0277248

            86. Mei T, Wang T, Deng Q, Gong Y. The Safety of Combining Immune Checkpoint Inhibitors and Platinum-Based Chemotherapy for the Treatment of Solid Tumors: A Systematic Review And Network Meta-Analysis. Frontiers in Immunology. 2023. Vol. 14:1062679

            87. Barroso-Sousa R, Barry WT, Garrido-Castro AC, Hodi FS, Min L, Krop IE, et al.. Incidence of Endocrine Dysfunction Following the Use of Different Immune Checkpoint Inhibitor Regimens: A Systematic Review and Meta-analysis. JAMA Oncology. 2018. Vol. 4:173–182

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            21 February 2024
            : 3
            : 1
            : 1-19
            Affiliations
            [a ]The First Hospital of China Medical University, Shenyang, Liaoning, China
            [b ]School of Pharmacy, China Medical University, Shenyang, Liaoning, China
            Author notes
            *Correspondence: liheranmm@ 123456163.com (H. Li); qhluo@ 123456cmu.edu.cn (Q. Luo)

            1Peipei Ouyang and Weiting Yang contributed equally to this work.

            Article
            10.15212/AMM-2023-0037
            015979e2-4f2e-44e5-9f85-5b7832fed323
            Copyright © 2024 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 12 September 2023
            : 02 January 2024
            : 18 January 2024
            Page count
            Figures: 7, Tables: 1, References: 87, Pages: 19
            Funding
            Funded by: National Natural Science Foundation of China
            Award ID: 81803442
            This study was supported by a grant from the National Natural Science Foundation of China (No. 81803442).
            Categories
            Review Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            Endocrine toxicity,Combination therapy,Network meta-analysis (NMA),ICI,Immune-related adverse events (irAEs)

            Comments

            Comment on this article