The Toll-like receptor (TLR) 7/8 agonist resiquimod has been used as an immune adjuvant in cancer vaccines. We evaluated the safety and immunogenicity of the cancer testis antigen NY-ESO-1 given in combination with Montanide (Seppic) with or without resiquimod in patients with high-risk melanoma. In part I of the study, patients received 100 μg of full-length NY-ESO-1 protein emulsified in 1.25 mL of Montanide (day 1) followed by topical application of 1,000 mg of 0.2% resiquimod gel on days 1 and 3 (cohort 1) versus days 1, 3, and 5 (cohort 2) of a 21-day cycle. In part II, patients were randomized to receive 100-μg NY-ESO-1 protein plus Montanide (day 1) followed by topical application of placebo gel [(arm A; n = 8) or 1,000 mg of 0.2% resiquimod gel (arm B; n = 12)] using the dosing regimen established in part I. The vaccine regimens were generally well tolerated. NY-ESO-1-specific humoral responses were induced or boosted in all patients, many of whom had high titer antibodies. In part II, 16 of 20 patients in both arms had NY-ESO-1-specific CD4⁺ T-cell responses. CD8⁺ T-cell responses were only seen in 3 of 12 patients in arm B. Patients with TLR7 SNP rs179008 had a greater likelihood of developing NY-ESO-1-specific CD8⁺ responses. In conclusion, NY-ESO-1 protein in combination with Montanide with or without topical resiquimod is safe and induces both antibody and CD4⁺ T-cell responses in the majority of patients; the small proportion of CD8⁺ T-cell responses suggests that the addition of topical resiquimod to Montanide is not sufficient to induce consistent NY-ESO-1-specific CD8⁺ T-cell responses.
e14010 Background: Polyinosinic-polycytidylic acid (poly-ICLC) is stabilized double stranded RNA comprising carboxymethylcellulose, polyinosinic-polycytidylic acid, and poly-L-lysine RNA. Poly-ICLC is a ligand for toll like receptor 3 (TLR3) expressed on dendritic cells and is under clinical development for the treatment of viral infections and tumors. Pilot studies in pediatric brain cancer have shown efficacy.1 Poly-ICLC can be given intra-muscularly (IM) to induce systemic inflammation and/or intratumorally (IT) to induce immune infiltration of tumor. Methods: Three patients are currently enrolled on this study. The planned sample size is 15. Eligible patients include those ≥18 years old with unresectable melanoma, squamous cell cancer of the head and neck, breast cancer, squamous cell cancer of the skin or basal cell cancer. Patients with sarcoma are also eligible and must be ≥14 years old. Patients must have an ECOG performance status of 2 or better, a life expectancy of 6 months and must have progressed on prior systemic therapy or despite local irradiation. Brain metastases are allowable. 1 mg Poly-ICLC is administered IT three times a week for two weeks followed by biweekly IM injection for 7 weeks and 1 week of rest. If stable disease or better is observed, this 10 week cycle will be repeated and if there is continuous response than a maintenance cycle of IM poly-ICLC will be offered. The primary objectives of the study are to determine safety and evaluate therapeutic effect as assessed by progression free survival at 26 weeks. The secondary objectives are to determine whether injected and/or non-injected lesions will regress and to assess immunogenicity of this poly-ICLC regimen. Results: Not yet available. Conclusions: Not yet available. Clinical Trial Information: NCT01984892 1. Hartman LL, Crawford JR, Makale MT, et al. Pediatric Phase II Trials of Poly-ICLC in the Management of Newly Diagnosed and Recurrent Brain Tumors. Journal of pediatric hematology/oncology 2013. Clinical trial information: NCT01984892.
166 Background: Poly-ICLC, a RNA complex, can activate dendritic cells and trigger NK cells to kill tumor cells. It can be given intramuscularly (IM) to induce systemic inflammation and intratumorally (IT) to induce immune infiltration of tumors. Methods: In this phase II study, eligible subjects are head and neck, and skin cancer patients with refractory or metastatic disease. In each treatment cycle, one accessible tumor site was targeted for IT injection of 1 mg of Poly-ICLC 3 times a week for 2 weeks followed by IM boosters biweekly for 6 weeks and then a 2 week rest period. Tumor biopsies were performed at baseline, week 3, and week 26. Pre- and post-vaccination tumors were evaluated by quantitative immunohistochemistry (IHC) and RNA sequencing. Blood samples were collected at baseline and prior to each cycle for immune response evaluations. Results: A phase I pilot portion of this study showed 1 patient who achieved clinical benefit with stable disease (progression-free survival of 6 months). Poly-ICLC was well tolerated with principal side effects of fatigue and inflammation at injection site (< grade 2). One case of grade 3 tumor necrosis was observed. In the patient with clinical benefit, IHC analysis of tumor showed increased CD4(60x), CD8(10x), PD-1(20x), and PD-L1(3x) compared to patients with progressive disease whose tumor biopsies showed unchanged/decreased CD4, CD8, PD-1, and PD-L1 levels over treatment period. Furthermore, RNASeq analysis of the same patient’s tumor and peripheral blood mononuclear cells (PBMC) showed dramatic changes such as upregulation of interferon-stimulated genes, chemokines, and genes associated with T cell activation and antigen presentation indicative of local and systemic immune activation in response to Poly-ICLC treatment. Conclusions: Preliminary findings show that Poly-ICLC is well tolerated in advanced solid cancer patients, and generates local immune response in tumor microenvironment and systemic immune response as evident in the patient achieving clinical benefit. These results warrant further investigation, and are currently being explored in this ongoing larger multicenter adaptive phase II clinical trial. Clinical trial information: NCT01984892.
Immunomodulation has shown great promise as an Alzheimer's disease (AD) therapy but major limitations must be overcome, such as the need for effectively reducing cerebral amyloid angiopathy (CAA), without associated hemorrhages. CAA is a common feature in AD and cognitively normal elderly individuals. Our initial findings indicate that stimulation of innate immunity with CpG ODN appears to be an effective means of reducing vascular amyloid without inducing toxicity in AD mouse models. We are currently testing our approach in a well characterized non-human primate model of sporadic CAA, squirrel monkey (Saimiri Boliviensis), which share numerous biological similarities with humans. Varying doses of the class B CpG ODN preparation containing the primate specific immunostimulatory sequence were administered in young monkeys by a subcutaneous route (s.c.). The most effective and non-toxic dosage described in young animals was selected for our long term studies in the older animals (with expected CAA deposits). During the treatment, primates were closely monitored for signs of toxicity. The peripheral cytokine responses were determined in PBMC supernatants and plasma from control and treated monkeys at selected time points. In addition, the performances on cognitive tests are being compared between our aged and young squirrel monkeys. We have shown that a TLR9 agonist prevented short-term memory deficits in Tg-SwDI mice, a model with extensive CAA. CpG ODN treatment led to a reduction of CAA in the absence of microhemorrhages and increased inflammation. Transgenic models are ideal for initial screening of a potential therapy but prior to clinical trials it is imperative to perform studies in non-human primates, which are a more biologically proximate model to humans. Short duration safety and efficacy assessment studies were first performed in young monkeys. Characterization of immune responses and evaluation of cognitive function and working memory in our monkeys is currently in progress. Assessment of the therapeutic potential of CpG ODN in aged squirrel monkeys, at a point where CAA is already present, is on-going. Overall, the studies described here provide a basis for potential future clinical trials of TLR9 stimulation with CpG ODNs to ameliorate AD related pathology in humans.
e20025 Background: Ipilimumab (Ipi) has improved clinical outcomes in metastatic melanoma (MM). Previous data suggested that low doses of cyclophosphamide (ldCTX) may have immunomodulatory effects including selectively decreasing T regulatory cells, down-regulating CTLA-4 on T cells, and altering cytokine profile. On this basis, we studied the combination of ldCTX and Ipi in patients (pts) with MM for overall response rates (RRs) and explored immunologic correlates. Methods: 10 pts with MM (2F:8M; median age 59; 7 cutaneous MM, 2 ocular MM and 1 unknown primary MM.) were enrolled in the stage 1 phase of the Simon 2 stage designed trial. Pts received induction tx with ldCTX (300 mg/m2) on day 1 followed by Ipi 10 mg/kg on day 3 for four cycles. Tumor assessments were done on Wk 12 and 24. Pts who showed clinical benefit at Wk 24 received ipi only maintenance tx for 3 cycles q 12 wks. Reassessment was on Wk 36, 48 and 60. Exploratory objectives including the T cell profile, antigen specific T cell responses, cytokine profile, CTLA-4 polymorphisms are being completed. Results: Six pts completed all 4 cycles of induction, and 4 pts received at least 2- 3 cycles. At Wk 24, 4 pts had stable disease, and 6 pts had progression of disease. Two pts received 1 cycle of maintenance tx. The 2 other SD pts were removed from study due to toxicity. Since the primary endpoint was objective response rate and none were seen, the trial was terminated based on trial design. 3 pts experienced Grade IV steroid refractory diarrhea that required anti-TNF alpha tx, and 1 patient experienced Grade III myopathy requiring a cholinesterase inhibitor for myasthenia gravis-like symptoms. Conclusions: Combination tx with ldCTX and ipi did not increase RRs; however did appear to significantly increase irAEs. Results of exploratory objectives may further elucidate mechanisms of enhanced toxicity and reduced efficacy. Clinical trial information: NCT01740401. irAE Total number of events Grade I Grade II Grade III-IV Grade III-IV irAE’s in Registration Trial (%) (Hodi, 2010NEJM) Rash 7 5 (50%) 2 (20%) 0 2.5% Diarrhea/colitis 6 0 3 (30%) 3 (30%) 4.6%/5.3% Nausea 1 1 0 0 2.3% Endocrinopathy 1 1 0 0 3.8% Myopathy/neuropathy 1 0 0 1 1.5%
Objective: Toll-like receptor-3 agonist Poly-ICLC has been known to activate immune cells and induce HIV replication in pre-clinical experiments. In this study we investigated if Poly-ICLC could be used for disrupting HIV latency while simultaneously enhancing innate immune responses. Design: This was a randomized, placebo-controlled, double-blinded trial in aviremic, cART-treated HIV-infected subjects. Participants (n=15) were randomized 3:1 to receive two consecutive daily doses of Poly-ICLC (1.4mg subcutaneously) versus placebo. Subjects were observed for adverse events, immune activation and viral replication. Methods: Besides primary outcomes of safety and tolerability, several longitudinal immune parameters were evaluated including immune cell phenotype and function via flowcytometry, ELISA and transcriptional profiling. PCR assays for plasma HIV-1 RNA, CD4+ T cell-associated HIV-1 RNA and proviral DNA were performed to measure HIV reservoirs and latency. Results: Poly-ICLC was overall safe and well-tolerated. Poly-ICLC-related adverse events were Grade 1/2, with the exception of one Grade 3 neutropenia which was short-lived. Mild Injection site reactions were observed in nearly all participants in the Poly-ICLC arm. Transcriptional analyses revealed upregulation of innate immune pathways in PBMCs following Poly-ICLC treatment, including strong interferon signaling accompanied by transient increases in circulating IP-10 (CXCL10) levels. These responses generally peaked by 24-48 hours after the first injection and returned to baseline by day 8. CD4+ T cell number and phenotype were unchanged, plasma viral control was maintained and no significant effect on HIV reservoirs was observed. Conclusions: These finding suggest that Poly-ICLC could be safely used for inducing transient innate immune responses in treated HIV+ subjects indicating promise as an adjuvant for HIV therapeutic vaccines. Trial Registration: ClinicalTrials.gov identifier: NCT02071095.
3084 Background: Cancer-testis antigen NY-ESO-1 is a highly immunogenic melanoma antigen which has been adopted for adjuvant vaccine trials. Three such trials (NCT00124124, NCT00821652, NCT01079741) in high-risk melanoma patients were conducted at our center. Although exact treatment regimens differed between trials, all enrolled patients received an NY-ESO-1-based peptide vaccine. Methods: All vaccine trial patients had histologically-proven melanoma and were without evidence of disease at trial entry; every stage III patient for whom records were available was included for analysis. For comparison, a control cohort of stage III patients who received no adjuvant therapy was identified via our clinicopathological database; every such patient in the database was included for analysis. Recurrence patterns were compared via a chi-squared test. Overall survival (OS) was estimated by Kaplan-Meier analysis, and a Cox proportional hazard model was employed to control for thickness and ulceration, and calculate a hazard ratio (HR). Results: Information was available for 62 vaccinated patients and 116 control patients (median follow-up: 62.8 and 30.5 months, respectively). The vaccine and control groups had the following characteristics, respectively: male (52% vs 62%), median thickness (2.0 vs 2.7), ulceration (41% vs 47%), positive lymph nodes (81% vs 93%), and nodular histotype (52% vs 56%); only lymph node status was significantly different (p = 0.02). At last follow-up, 35 patients (56%) in the treatment group had recurred, compared to 84 patients (64%) in the control group. At first recurrence, vaccinated patients tended to recur with resectable disease more often than control patients (79% vs 60%, p = 0.10), though this result was not significant. OS, however, was significantly longer in the vaccine group compared to the control group (median not reached vs 57.5 months, HR 0.58, 95%CI 0.34 – 0.99, p = 0.047). Conclusions: In this small retrospective cohort of stage III melanoma patients, adjuvant NY-ESO-1 vaccination was associated with improved OS. NY-ESO-1 vaccination may exert an immunologic advantage reflected in a pattern of locally resectable disease upon recurrence compared to diffusely metastatic recurrences.