scholarly journals Modulation of temozolomide dose differentially affects T-cell response to immune checkpoint inhibition

2019 ◽  
Vol 21 (6) ◽  
pp. 730-741 ◽  
Author(s):  
Aida Karachi ◽  
Changlin Yang ◽  
Farhad Dastmalchi ◽  
Elias J Sayour ◽  
Jianping Huang ◽  
...  

Abstract Background The changes induced in host immunity and the tumor microenvironment by chemotherapy have been shown to impact immunotherapy response in both a positive and a negative fashion. Temozolomide is the most common chemotherapy used to treat glioblastoma (GBM) and has been shown to have variable effects on immune response to immunotherapy. Therefore, we aimed to determine the immune modulatory effects of temozolomide that would impact response to immune checkpoint inhibition in the treatment of experimental GBM. Methods Immune function and antitumor efficacy of immune checkpoint inhibition were tested after treatment with metronomic dose (MD) temozolomide (25 mg/kg × 10 days) or standard dose (SD) temozolomide (50 mg/kg × 5 days) in the GL261 and KR158 murine glioma models. Results SD temozolomide treatment resulted in an upregulation of markers of T-cell exhaustion such as LAG-3 and TIM-3 in lymphocytes which was not seen with MD temozolomide. When temozolomide treatment was combined with programmed cell death 1 (PD-1) antibody therapy, the MD temozolomide/PD-1 antibody group demonstrated a decrease in exhaustion markers in tumor infiltrating lymphocytes that was not observed in the SD temozolomide/PD-1 antibody group. Also, the survival advantage of PD-1 antibody therapy in a murine syngeneic intracranial glioma model was abrogated by adding SD temozolomide to treatment. However, when MD temozolomide was added to PD-1 inhibition, it preserved the survival benefit that was seen by PD-1 antibody therapy alone. Conclusion The peripheral and intratumoral immune microenvironments are distinctively affected by dose modulation of temozolomide.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4918-4918
Author(s):  
Rachel Cantrell ◽  
Leah A. Rosenfeldt ◽  
Duaa Mureb ◽  
Balkrishan Sharma ◽  
Alexey Revenko ◽  
...  

A serious and life-threatening cancer-associated sequelae is venous thromboembolism (VTE). Indeed, VTE is the second leading cause of death in cancer patients, second only to the malignancy itself. Cancer patients with VTE or at high risk for VTE are generally treated with anticoagulants, which limit thrombin generation. While it's been widely accepted that thrombin plays a role in cancer progression, the effects thrombin has on other cell types within the tumor microenvironment (TME) have not been thoroughly studied. An understudied role of thrombin may be found in its ability to drive T cell functions. Recently, we have identified thrombin as a potential enhancer of CD8+ T cell effector functions by signaling through the protease activated receptor 1 (PAR-1). Our preliminary data shows that thrombin increases CD8+ T cell survival in a PAR-1 dependent fashion. CD8+ tumor infiltrating lymphocytes (TILs) play a critical role in tumor clearance through their cytolytic and anti-tumor cytokine producing capacity. However, the hostile tumor microenvironment (TME) promotes a gradual reduction in CD8+ TIL capacity to produce cytokines and kill targets. Specific components of the TME, including PDL1 expression, are associated with loss of T cell functionality. A promising strategy to block the interaction of CD8+ TILs and the inhibitory TME components is immune checkpoint inhibition (ICI) therapy, as shown by effective blockade of PD1 signaling by anti-PD1 antibodies. However, these ICI therapies leave many patients unresponsive, highlighting the necessity to uncover additional underlying mechanisms involved in modulating CD8+ T cell responses against cancer. Our preliminary findings lead us to hypothesize that thrombin, in conjunction with PD1 blockade, may work in synergy to promote CD8+ T cell killing of tumors. Consistent with our hypothesis, preliminary results suggest that thrombin is necessary for a robust anti-tumor immune response following ICI in vivo. Here, cohorts of C57BL/6 mice with low or normal circulating prothrombin levels bearing B16 tumors were treated with an anti-PD1 antibody or control IgG. Anti-PD1 therapy significantly limited tumor growth in mice with normal prothrombin levels, but had no impact on tumor growth in mice with low prothrombin levels. A major implication of our findings is that limiting thrombin generation with anticoagulants may be detrimental in the context of immune checkpoint inhibition treatment. Better defining the potential risk of reducing ICI efficacy by concurrent treatment with anticoagulants will require a detailed understanding of the role thrombin plays in cancer immunobiology. Disclosures Revenko: Ionis Pharmaceuticals: Employment. Monia:Ionis Pharmaceuticals: Employment. Palumbo:Ionis Pharmaceuticals: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1666-1666 ◽  
Author(s):  
Amer M. Zeidan ◽  
Joshua F. Zeidner ◽  
Amy Duffield ◽  
Hanna A. Knaus ◽  
Anna Ferguson ◽  
...  

Abstract Background: Patients (pts) with higher-risk (HR)-MDS in whom HMAs fail have a dismal prognosis with a median overall survival (OS) of <6 months. Immune tolerance and evasion by malignant cells have been recognized as important mechanisms of progression in cancers including MDS. Immune checkpoint inhibition with the anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) antibody ipilimumab improved outcomes in pts with solid tumors by overcoming this resistance mechanism. We hypothesized that CTLA-4 blockade in pts with HR-MDS post HMAs failure would be tolerable and lead to meaningful clinical responses. Methods: In an investigator-initiated, CTEP-sponsored phase 1b study, eligible pts with HR-MDS (defined by Intermediate-2 or high IPSS score, or intermediate-1 with ≥5% blasts in bone marrow [BM] or transfusion needs) who had primary or secondary failure of HMAs (≥4 cycles) received ipilimumab monotherapy at 2 dose levels (DL); DL1 used 3mg/kg and DL2 used 10mg/kg. In the induction phase [Figure 1], 4 doses were administered at 3-week intervals. Responding Pts and those with stable disease (SD) at end of induction received a maintenance phase with 4 more doses of ipilimumab administered at 3-month intervals (study total is 8 doses). BM biopsies were obtained at baseline, post cycles 2, 4, and before each maintenance dose. The primary objectives were to determine the tolerability of ipilimumab and identify the optimal dose for dose expansion. Toxicities were graded according to CTCAE 4.0 criteria. Responses were evaluated using International Working Group 2006 criteria at end of induction and before each maintenance dose. Overall survival (OS) was calculated from first dose of ipilimumab until death using Kaplan-Meier methods with censoring at time of allogeneic stem cell transplantation (alloSCT) or cutoff date for data collection on 6/30/2015. Results: Eleven pts were enrolled on the dose-escalation part of the study; 6 on DL1 and 5 on DL2. Median age was 63 years (range, 50-79). Five pts had received 2 prior lines of therapy (including HMAs) while 6 pts had received only HMAs; median number of HMAs cycles 5; range, 4-18. Five pts had a normal karyotype while each of the following categories were present in one pt: del5q, del7q, complex, trisomy 8 (2 pts unsuccessful karyotyping). At baseline, 4 pts were platelet transfusion-dependent and 5 were red blood cell transfusion-dependent. Median platelet count was 25×10⁹/L (range, 12-61×10⁹/L), median white blood cell count was 1.5×10⁹/L (range, 1.05-7.6×10⁹/L), median absolute neutrophil count was 0.28×10⁹/L (range, 0.06-4.7×10⁹/L), median hemoglobin was 9.4 gm/dL (range 7.6-10.8 gm/dL), and median BM blast percentage was 9% (range, 2-30%). All pts in DL1 received ≥2 ipilimumab doses; 2 received 6 or 8 doses. Three of 6 pts in DL1 developed grade 3 immune-related adverse events (IRAEs) while 4 of 5 pts at DL2 developed grade 3 IRAEs. Four pts (36%) experienced no IRAEs (3 at DL1 and 1 at DL2). All IRAEs were reversible with stopping ipilimumab +/- initiation of systemic steroids. No pt died due to an IRAE and all patients were successfully weaned from steroids. The spectrum of IRAEs was similar to that of ipilimumab use in pts with solid malignancies (rash, hepatitis, and diarrhea/colitis). Overall no pts had objective responses. Three pts (27.3%) had SD for >6 months; one had a prolonged SD after an IRAE, and one experienced an ongoing SD for >16 months. Three pts underwent alloSCT post ipilimumab without any additional toxicities and remain in complete remission at 2, 12 and 18 months post alloSCT, respectively. Median and mean OS for entire cohort (censoring at time of alloSCT) was 368 and 352 days, respectively (95%CI for mean OS, 264-440 days) [Figure 2]. Correlative studies evaluating dynamic changes in T-cell subsets, myeloid derived suppressor cells, cytokine levels, and T-cell receptor repertoire are in progress. Conclusions: Monotherapy withanti-CTLA-4 antibody ipilimumabat 3mg/kg is tolerable and can lead to prolonged disease stabilization. This dose is currently being evaluated further in a dose-expansion multi-center study. AlloSCT appears feasible post ipilimumab use but further data are needed. These results provide rationale for further exploration of immune checkpoint inhibition therapy in pts with MDS. Figure 1. Administration schedule Figure 1. Administration schedule Figure 2. Overall survival from first dose (censored at alloSCT or 6/30/2015). Figure 2. Overall survival from first dose (censored at alloSCT or 6/30/2015). Disclosures Off Label Use: Ipilimumab for MDS. Gore:Celgene: Consultancy, Honoraria, Research Funding.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A311-A311
Author(s):  
Alexander Chacon ◽  
Alexa Melucci ◽  
Shuyang Qin ◽  
Paul Burchard ◽  
Katherine Jackson ◽  
...  

BackgroundOnly 30–40% of metastatic melanoma patients experience objective responses to first line anti-PD-1 immune checkpoint inhibition (αPD-1 ICI). Cyclooxygenase (COX-1/2) inhibition with aspirin (ASA) and other non-steroidal anti-inflammatory drugs has been associated with prolonged time to recurrence and improved responsiveness to ICI in human melanoma,1 with inhibition of myeloid-induced immunosuppression in the tumor microenvironment (TME) a purported mechanism.2 Similarly, dietary omega-3 fatty acids metabolized by COX-2 elicit downstream effects on T-cell differentiation akin to ASA administration, abrogating murine melanoma and human breast cancer progression. Mechanisms of ICI resistance remain unclear, and adjunct therapies look to bridge the gap from current response rates to cure.MethodsYUMM 1.7 melanoma cells were injected into flanks of C57-BL6/J mice. Mice were fed control diets or supplemented with omega-3 rich fish oil (FO) chow (10% weight/weight, 30%kcal/kcal), ASA in drinking water (ASA, LO – 300, MED – 600, HI - 1000 ug/mL), or the combination of these agents (COMBO, with ASA-MED) starting at day 7 post tumor implantation. Intraperitoneal αPD1 was administered every 3–4 days starting at day 12. Tumors were assessed for growth, harvested at day 32 (day 26 for ASA LO/HI), and characterized with flow cytometry. All significant results (p<0.05) assessed by 2-way ANOVA or t-test as appropriate.ResultsFO resulted in lesser tumor volume at day 32 in αPD-1 treated mice, while ASA-HI resulted in lesser tumor volume in mice not treated with αPD-1 but did not synergize with αPD-1. ASA-MED and COMBO groups trended towards decreased tumor size (p = 0.07 and 0.07 respectively) by day 32 in αPD-1 treated mice. FO and COMBO increased total CD3+ T-cells and monocytes (CD45+, CD19-, CD11b+, Ly6C+, Ly6G -) in the TME. FO increased PD-L1 + CD4+ T-cells, while COMBO increased total CD8+ T-cells and PD1+ CD8+ T-cells. ASA-HI increased monocytes and the proportion of PD-1+, CD8+ T-cells in the TME.ConclusionsMyeloid-induced suppression of T-cell function in tumors may contribute to immune checkpoint inhibition resistance. In the present study, both fish oil and aspirin altered melanoma tumor growth, with only fish oil synergizing with anti-PD-1 at the doses assessed. Both fish oil and aspirin augmented monocyte populations in the tumor microenvironment, with differential effects on T-cell populations. The partially synergistic mechanism between substrate-limited (FO) and pharmacologic (ASA) inhibition of cyclooxygenase-2 may provide a cost-effective avenue to combat immune escape in melanoma patients treated with anti-PD-1 immune checkpoint inhibition, requiring further investigation in humans.ReferencesWang SJ, et al. Effect of cyclo-oxygenase inhibitor use during checkpoint blockade immunotherapy in patients with metastatic melanoma and non-small cell lung cancer. J Immunother Cancer 2020;8(2).Zelenay S, et al. Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell 2015;162(6):1257–70.


2017 ◽  
Author(s):  
Valeria Quaranta ◽  
Carolyn Rainer ◽  
Sebastian R. Nielsen ◽  
Meirion Raymant ◽  
Muhammad Shamsher Ahmed ◽  
...  

AbstractThe ability of disseminated cancer cells to evade the immune response is a critical step for efficient metastatic progression. Protection against an immune attack is often provided by the tumour microenvironment that suppresses and/or excludes cytotoxic CD8+ T cells. Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive metastatic disease with unmet needs, yet the immuno-protective role of the metastatic tumour microenvironment in pancreatic cancer is not completely understood. In this study we find that macrophage-derived granulin contributes to cytotoxic CD8+ T cell exclusion in metastatic livers. Mechanistically, we find that granulin expression by macrophages is induced in response to colony stimulating factor-1. Genetic depletion of granulin reduces the formation a fibrotic stroma, thereby allowing T cell entry at the metastatic site. While metastatic PDAC tumours are largely resistant to anti-PD-1 therapy, blockade of PD-1 in granulin depleted tumours restored the anti-tumour immune defence and dramatically decreased metastatic tumour burden. These findings suggest that targeting granulin may serve as a potential therapeutic strategy to restore CD8+ T cell infiltration in metastatic PDAC, thereby converting PDAC metastatic tumours, which are refractory to immune checkpoint inhibitors, into tumours that respond to immune checkpoint inhibition therapies.


Sign in / Sign up

Export Citation Format

Share Document