DC-HIL/Gpnmb Is a Negative Regulator of Tumor Response to Immune Checkpoint Inhibitors

2019 ◽  
Vol 26 (6) ◽  
pp. 1449-1459 ◽  
Author(s):  
Jin-Sung Chung ◽  
Vijay Ramani ◽  
Masato Kobayashi ◽  
Farjana Fattah ◽  
Vinita Popat ◽  
...  
2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e16528-e16528
Author(s):  
Liping Li ◽  
Mengmei Yang ◽  
Mengli Huang

e16528 Background: Immune checkpoint inhibitors (ICIs) targeting PD-1/L1 have been approved as first-line treatment for cisplatin-ineligible patients and as second-line therapy for patients with metastatic urothelial carcinoma of the bladder. Biomarkers can help select patients who are more likely to response to ICIs. RNF43 is an E3 ubiquitin ligase that acts as a negative regulator of Wnt/β-catenin signaling pathway. In colorectal cancer (CRC) patients treated with immune checkpoint inhibitors (ICIs), RNF43 mutations predicted longer overall survival (OS). The impact of RNF43 mutations on the efficiency of ICIs in bladder cancer(BLC) remains to be explored. Methods: We downloaded the mutation and clinical data of 211 BLC patients treated with ICIs from the immunotherapeutic cohort published by Samstein et al. (2019). OS analyses were conducted using Kaplan-Meier curves and log-rank tests. Wilcoxon test was used for the comparison of TMB. We also downloaded a TCGA cohort for prognostic analysis. The correlations between RNF43 and immune infiltrates were analyzed in the TIMER2.0 database. Statistical significance was set at p = 0.05. Results: RNF43 mutations were identified in 4.3%(9/211) and 3%(13/438) BLC patients in the immunotherapeutic and TCGA cohort, respectively. In the immunotherapeutic cohort, patients with RNF43 mutations had significantly longer OS (25 months vs 8 months; p = 0.015) and higher tumor mutation burden(TMB, 42.3 vs 7.9; p = 3.15E-06) than RNF43-wild-type patients. Different from this, no significant difference was found in OS between RNF43-mutant and RNF43-wild-type BLC patients with standard treatment in the TCGA cohort (p = 0.696). These results indicated that RNF43 was not a prognostic factor but a predictive biomarker of survival in BLC treated with ICIs. No difference was observed in subsets of immune cells between RNF43-mutant and the RNF43-wide-type BLC patients, including neutrophils, macrophages, CD8+ T cells, Tregs, B cells and NK cells. Conclusions: RNF43 mutations may be a predictor of survival benefit from ICIs in bladder cancer and correlated with higher TMB. Further studies in other ICI-treated cohorts are needed to confirm these results.


2021 ◽  
Vol 11 ◽  
Author(s):  
Yong Fan ◽  
Wenhui Xie ◽  
Hong Huang ◽  
Yunxia Wang ◽  
Guangtao Li ◽  
...  

ObjectivesImmune checkpoint inhibitors (ICIs) have brought impressive benefits to cancer patients, however often accompanied with immune-related adverse events (irAEs). We aimed to investigate the association of irAEs with efficacy and overall survival in cancer patients treated by ICIs, and further quantify the association by stratifying subgroups.MethodsPubMed, EMBASE and Cochrane library from database inception to 29 August 2019 were systematically searched. Articles reporting association of objective response rate (ORR), progression-free survival (PFS), overall survival (OS) with irAEs in cancer patients treated with approved ICIs were included. Adjusted odds ratios (OR) with 95% confidential intervals (CIs) were calculated for ORR, and hazard ratios (HR) were used for PFS and OS.ResultsA total of 52 articles comprising 9,156 patients were included. Pooled data demonstrated a statistically significant greater probability of achieving objective tumor response for patients with irAEs compared to those without (OR 3.91, 95% CI 3.05–5.02). In overall meta-analysis, patients who developed irAEs presented a prolonged PFS (HR 0.54; 95% CI 0.46–0.62) and OS (HR 0.51; 95% CI 0.41–0.59). More specifically, irAEs in certain cancer types (NSCLC and melanoma) and organs (skin and endocrine) were robustly associated with better clinical outcomes, while this association needs further verification regarding other tumors. High grade toxicities (G3–5) were not associated with a significantly favorable PFS or OS. Additionally, the association between irAEs and clinical benefit seemed to be more definite in patients receiving PD-(L)1 blockade than CTLA-4 blockade. Pooled data from landmark analyses displayed consistent results.ConclusionsThe occurrence of irAEs predicted improved tumor response and better survival in overall cancer patients treated with ICIs. Notably, the association stayed robust in certain cancer types (NSCLC and melanoma) and organ-specific irAEs (skin and endocrine).


Liver Cancer ◽  
2019 ◽  
Vol 8 (6) ◽  
pp. 480-490 ◽  
Author(s):  
Li-Chun Lu ◽  
Chiun Hsu ◽  
Yu-Yun Shao ◽  
Yee Chao ◽  
Chia-Jui Yen ◽  
...  

Cancers ◽  
2021 ◽  
Vol 13 (15) ◽  
pp. 3738
Author(s):  
Marzia Del Re ◽  
Caterina Vivaldi ◽  
Eleonora Rofi ◽  
Francesca Salani ◽  
Stefania Crucitta ◽  
...  

Pancreatic ductal adenocarcinoma (PDAC) is a non-immunogenic tumor poorly responsive to immune checkpoint inhibitors. This study investigates the effect of 5-fluorouracil (5-FU), irinotecan, and oxaliplatin (FOLFIRINOX), and gemcitabine plus nab-paclitaxel (GEMnPAC) regimens on PD-L1 mRNA expression in plasma-derived microvesicles (MVs) in 50 PDAC patients. Plasma was collected before starting chemotherapy and after 3 months of treatment. mRNA was extracted from MVs, and PD-L1 expression was measured by digital droplet PCR. Twenty-eight patients were PD-L1 positive in MVs at baseline, of which 18 were in the GEMnPAC cohort and 10 in the FOLFIRINOX one. The amount of PD-L1 expression in MVs increased from baseline to 3 months of treatment in patients receiving GEMnPAC (median value 0.002 vs. 0.005; p = 0.01) compared to those treated with FOLFIRINOX (median 0.003 vs. 0.004; p = 0.97). The increase in PD-L1 mRNA expression in MVs was not related to tumor response (PR + SD: p = 0.08; PD: p = 0.28). Our findings demonstrate that GEMnPAC can increase PD-L1 mRNA expression in patient-derived circulating MVs, providing a rationale for testing the efficacy of this regimen in sequential or simultaneous combinations with immunotherapy in PDAC patients.


Author(s):  
H. M. Stege ◽  
M. Haist ◽  
S. Schultheis ◽  
M. I. Fleischer ◽  
P. Mohr ◽  
...  

Abstract Background Immune checkpoint inhibitors (ICI) have led to a prolongation of progression-free and overall survival in patients with metastatic Merkel cell carcinoma (MCC). However, immune-mediated adverse events due to ICI therapy are common and often lead to treatment discontinuation. The response duration after cessation of ICI treatment is unknown. Hence, this study aimed to investigate the time to relapse after discontinuation of ICI in MCC patients. Methods We analyzed 20 patients with metastatic MCC who have been retrospectively enrolled at eleven skin cancer centers in Germany. These patients have received ICI therapy and showed as best overall response (BOR) at least a stable disease (SD) upon ICI therapy. All patients have discontinued ICI therapy for other reasons than disease progression. Data on treatment duration, tumor response, treatment cessation, response durability, and tumor relapse were recorded. Results Overall, 12 of 20 patients (60%) with MCC relapsed after discontinuation of ICI. The median response durability was 10.0 months. Complete response (CR) as BOR to ICI-treatment was observed in six patients, partial response (PR) in eleven, and SD in three patients. Disease progression was less frequent in patients with CR (2/6 patients relapsed) as compared to patients with PR (7/11) and SD (3/3), albeit the effect of initial BOR on the response durability was below statistical significance. The median duration of ICI therapy was 10.0 months. Our results did not show a correlation between treatment duration and the risk of relapse after treatment withdrawal. Major reasons for discontinuation of ICI therapy were CR (20%), adverse events (35%), fatigue (20%), or patient decision (25%). Discontinuation of ICI due to adverse events resulted in progressive disease (PD) in 71% of patients regardless of the initial response. A re-induction of ICI was initiated in 8 patients upon tumor progression. We observed a renewed tumor response in 4 of these 8 patients. Notably, all 4 patients showed an initial BOR of at least PR. Conclusion Our results from this contemporary cohort of patients with metastatic MCC indicate that MCC patients are at higher risk of relapse after discontinuation of ICI as compared to melanoma patients. Notably, the risk of disease progression after discontinuation of ICI treatment is lower in patients with initial CR (33%) as compared to patients with initial PR (66%) or SD (100%). Upon tumor progression, re-induction of ICI is a feasible option. Our data suggest that the BOR to initial ICI therapy might be a potential predictive clinical marker for a successful re-induction.


Author(s):  
Satoshi Furune ◽  
Chiaki Kondo ◽  
Yuko Takano ◽  
Tomoya Shimokata ◽  
Mihoko Sugishita ◽  
...  

AbstractVitiligo, an acquired depigmenting disorder of the skin that reacts against normal melanocytes, sometimes occurs as an immune-related adverse event in the treatment of melanoma with immune checkpoint inhibitors. It has been known that the occurrence of vitiligo is associated with a favorable therapeutic response in patients with melanoma, but it is not yet clear whether the association also applies to amelanotic melanoma, a minor subtype of melanoma with little or no melanin pigmentation. We report a patient with amelanotic melanoma of the esophagus who responded well to nivolumab treatment. Shortly after the tumor response, vitiligo was found on the patient’s forearms. This case suggests that the occurrence of vitiligo is associated with a favorable response to nivolumab treatment for amelanotic melanoma.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1552-1552
Author(s):  
Hebb Jonathan ◽  
Holbrook E Kohrt

Abstract Immunotherapy with agents such as immune checkpoint inhibitors and tumor necrosis factor (TNF) receptor superfamily agonists is a groundbreaking development with proven benefits and great potential in the treatment of solid tumors and hematologic malignancies. Combination immunotherapy creates even more potential for efficacy by targeting synergistic immune pathways. However, systemically administered immunomodulators can activate T-cells non-specifically resulting in off-target toxicity, which is dose-limiting and potentially lethal in combination. Intratumoral administration of immunotherapeutic agents has several advantages including 1) higher concentrations of agents in close proximity to target antigens and tumor-infiltrating lymphocytes 2) lower doses overall with less systemic exposure and toxicity 3) potentially a novel mechanism of action such as depletion of intratumoral Tregs 4) potential for an abscopal effect, essentially acting as an in-situ cancer vaccine. Here we report a novel combination of immunomodulators, anti-CTLA4, CD137 and OX40 administered by intratumoral route in a mouse lymphoma model (A20) as well as a colon cancer model (MC38). Additionally, anti-PD1 and CpG were used as adjunctive therapies in the lymphoma model. CTLA4 and PD1 are immune checkpoint inhibitors, CD137 and OX40 are members of the TNF receptor superfamily, and CpG is a TLR9 agonist. For the A20 model, 2 tumors were created by injecting 1 x 10^7 A20 cells on each flank of BalbC mice. Treatment was started on day 8 when there were visible tumors and intratumoral injection was to the left tumor only. Groups included 1) PBS 2) triple combination of immunomodulators (anti-CTLA4, anti-CD137 and anti-OX40) at 30 mg each in 50 ml volume x 4 biweekly doses 3) triple combination x 2 biweekly doses followed by 2 doses of anti-PD1 at 150 mg in 100ul given intraperitoneally 4) anti-PD1 x 2 biweekly doses followed by 2 doses of triple combination and 5) CpG 50 mg concurrent with triple combination. For the MC38 model, 5 x 10^5 cells were injected bilaterally on the flanks of C57BL/6 mice. Mice were treated with either PBS or triple combination 3x/week for 6 doses and intratumoral injection was to the left tumor only. For the A20 model, all treatment groups had a significant anti-tumor response compared to PBS with systemic regressions including both the treated and untreated tumor. Complete response was observed in 100% of the triple combination group, 100% of the CpG and triple combination group, and in 86% of the triple combination followed by PD1 group. Group 3, PD1 followed by triple combination, tumors initially progressed with PD1 alone, albeit slower than untreated, then regressed with triple combination treatment with 57% ultimately tumor-free. For the MC38 model, the triple combination resulted in a systemically effective anti-tumor response with near complete regression of treated and untreated sites and a significant survival advantage. Thus we demonstrate this novel combination of immunomodulators delivered intratumorally induces a dramatic anti-tumor effect. Anti-PD1 given prior to the combination appears to abrogate the response. Overall these preliminary results of intratumoral administration of combination anti-CTLA4, CD137 and OX40, all agents in clinical trials, are promising for future clinical development via this safe and highly efficacious route of administration. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 2 (11) ◽  
pp. 2202-2207 ◽  
Author(s):  
H. M. Stege ◽  
F. Bradfisch ◽  
M. I. Fleischer ◽  
P. Mohr ◽  
S. Ugurel ◽  
...  

AbstractSignificant progress has been made in the treatment of advanced Merkel cell carcinoma (MCC) by establishing immune checkpoint inhibitors (ICI). Tumor progression, durable response, or adverse events may lead to ICI discontinuation in MCC patients. If in these patients tumor progression occurs, the question remains if re-induction with ICI achieves renewed tumor response. This retrospective multicenter study evaluated patients in with re-induction of anti-PD-1/anti-PD-L1 therapy for advanced MCC. Clinical data were extracted at treatment initiation, tumor response, treatment cessation, and subsequent tumor response to re-induction. Eight patients from seven centers (mean age 67.8 years) were included. The median duration of initial therapy with anti-PD-1/anti-PD-L1 was 9.6 months (2–21 months). Two patients achieved complete response (CR), four patients partial response (PR), one patient stable disease (SD), while in one patient progressive disease (PD) occurred as best overall response (BOR) to ICI. Reason for discontinuation of ICI was PD in three patients and severe adverse events in five patients. Following a median anti-PD-1/anti-PD-L1 therapy-free interval of 9.5 months (3–18 months), re-induction with ICI therapy was initiated. Five of eight patients (62.5%) achieved an objective response upon re-induction, while in three patients, no response could be observed. Notably, adverse events, which had led to the discontinuation of the first ICI treatment line, were not observed upon re-induction. The initial response to immune checkpoint inhibitors seems to be an important marker for successful re-induction. Interestingly, adverse events leading to treatment discontinuation were not observed during re-induction.


Sign in / Sign up

Export Citation Format

Share Document