349 Early safety and efficacy of a phase 1/2 open-label, multi-center trial of SNS-301 added to pembrolizumab in patients with advanced squamous cell carcinoma of the head and neck (SCCHN)

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A375-A375
Author(s):  
Alain Algazi ◽  
William Smith ◽  
Timothy Panella ◽  
Dong Shin ◽  
Marie-Louise Fjaellskog ◽  
...  

BackgroundEfficacy of anti-PD-1 therapy is attributed to the presence of infiltrating antigen-specific CD8+ T-cells. Despite the success of anti-PD-1 therapy, many patients with SCCHN present with immune desert or immune excluded tumors and only 13–18% of patients achieve tumor reductions. Given this low response rate, it is imperative to combine agents that generate or expand anti-tumor T cells, such as vaccines, with anti-PD-1 therapies. SNS-301 is a first-in-class, bacteriophage-based immune activating agent targeting human aspartate β-hydroxylase (ASPH), a tumor associated antigen overexpressed in multiple tumor types. SNS-301 is a self-adjuvanted vaccine consisting of λ-bacteriophage engineered to express an immunogenic fragment of ASPH fused to the phage gpD coat protein, previously shown to be well tolerated and generate an immune response (Phase 1, NCT03120832). The objectives of this trial are to evaluate safety, immunogenicity and preliminary efficacy of SNS-301 in combination with pembrolizumab in patients that did not achieve tumor reductions on anti-PD-1/PD-L1 therapy alone.MethodsThe study consists of an initial safety-run-in followed by a two-stage design. SNS-301 is delivered intradermally in addition to pembrolizumab in up to 30 patients with locally advanced unresectable or metastatic/recurrent SCCHN. Patients must have actively received anti-PD-1 therapy for ≥12 weeks, with a best response of stable disease (SD) or unconfirmed progressive disease (PD) per iRECIST. Patients provide pre, on-treatment and biopsies at PD (optional) to characterize the tumor microenvironment using NanostringTM, multiplex immunohistochemistry, and correlate with clinical outcomes. Blood samples are collected to evaluate T cell responses using flow cytometry, ELISA, ELISPOT.ResultsAs of July 23, 2020, 9 patients were enrolled. Median duration of ongoing anti-PD therapy was 37 weeks (range 20–101). The combination was well-tolerated with no DLTs and mostly Grade 1–2 unrelated adverse events. Two Grade 3 events were reported: hypertension (not related) and dehydration (related), the later reported as serious adverse event. Of seven patients eligible for efficacy analysis, one patient with PD-L1 negative disease had a partial response with a reduction of 29% at week 6 with deepening of the response to 43% at week 12 and one patient with progressive disease at study entry had stabilization of disease at week 6 and 12. Another two patients had stable disease for 30+ weeks and three patients had PD. Additional efficacy and immunological analyses are ongoing.ConclusionsEarly data show that the combination of SNS-301 and pembrolizumab has manageable toxicity and capacity to achieve long-term disease stability and objective tumor responses.Trial RegistrationNCT04034225Ethics ApprovalThis study has been approved by WIRB (20190628) as well as several institutional IRBs.

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 6029-6029
Author(s):  
Alain Patrick Algazi ◽  
Dhaval Shah ◽  
William Smith ◽  
Timothy J. Panella ◽  
Dong Moon Shin ◽  
...  

6029 Background: The absence of infiltrating antigen-specific CD8+ T-cells at baseline is associated with low response rates to PD-1 blockade. SCCHN tumors often exclude effector T cells, and 2nd line response rates are low (13-18%). Highly immunogenic, antigen specific antitumor vaccines may expand intratumoral CD8+ T cells, potentially increasing durable response rates to PD-1 blockade. SNS-301 is a first-in-class, bacteriophage-based immune activating agent targeting human aspartate β-hydroxylase (ASPH), a tumor associated antigen overexpressed in multiple tumor types. SNS-301 is a self-adjuvanted vaccine consisting of λ-bacteriophage engineered to express an immunogenic fragment of ASPH fused to the phage gpD coat protein. The study objectives are to evaluate safety, immunogenicity and preliminary efficacy of SNS-301 added to pembrolizumab in patients (pts) not achieving tumor reductions on PD-1 blockade alone. Methods: Intradermal SNS-301 was combined with pembrolizumab in pts with locally advanced unresectable (LA) or metastatic/recurrent (met) SCCHN with a best response of stable disease (SD) or unconfirmed progressive disease (uPD) on ongoing PD-1 blockade > 12 weeks. Pts provided pre and on-treatment biopsies to characterize the tumor microenvironment using Nanostring and multiplex immunohistochemistry (mIHC). Blood samples were collected to evaluate B and T cell responses using ELISA/ELISPOT assays. Results: As of February 4, 2021, 13 pts were enrolled. Median duration of PD-1 blockade was 48 weeks (range 14-114) at study entry. There were no DLTs & mostly Grade 1-2 unrelated adverse events. Only two related Grade 3 events were reported: rash & dehydration (also a serious adverse event). Ten pts were evaluable for efficacy: 1 pt with PD-L1 negative (neg) disease & SD on pembrolizumab monotherapy achieved a partial response (PR; -52% at 8 months), 4 pts achieved SD & 5 pts had progressive disease. Two of the pts with SD had long-lasting duration (8 & 10 months) of which the latter had PD-L1 neg disease. One pt with uPD at enrollment achieved SD for 4 months. Analyses of pre- & on-treatment biopsies from the PR pt demonstrated an increase in infiltrating CD8+ T cells, PD-L1 expression & PD-1/PD-L1 proximity measures. Nanostring analysis demonstrated increased gene expression signatures for immune cells in the PR pt that was concordant with the mIHC & clinical outcome. Conclusions: The combination of SNS-301 and pembrolizumab was well-tolerated and resulted in encouraging clinical efficacy in pts not expected to respond to PD-1 blockade alone. Translational data suggest cellular response to SNS-301 and transformation of a poorly inflamed tumor to an immunologically active tumor in a responding pt (PR). Based on these data, an additional cohort will start enrolling PD-1 blockade naïve pts with LA/met SCHNN in the front-line setting. Clinical trial information: NCT04034225.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 11515-11515
Author(s):  
Margaret von Mehren ◽  
Sujana Movva ◽  
Elizabeth A. Handorf ◽  
Priscilla Merriam ◽  
Jeffrey A. Morgan ◽  
...  

11515 Background: Dedifferentiated liposarcoma (DDL) is characterized by ring chromosomes of chromosone12, which includes amplification of MDM2 and CDK4. Exposure to CDK4 inhibitors in Rb+ leiomyosarcoma (LMS) cell lines leads to decreased cell proliferation, and increased senescence, and G0/G1-phase arrest. When combined, ribociclib a CDK4 inhibitor and everolimus, an mTOR inhibitor show synergistic growth inhibition in multiple tumor models. We hypothesized that this combination could lead to increased disease control in patients with DDL. Methods: This study enrolled patients (pts) into one of two cohorts: DDL or Rb+ LMS. LMS pts were required to have 1 prior line of therapy; DDL pts required no prior therapy. There were no limits to prior therapies in either group. Measurable disease by RECIST 1.1 was also required. Ribociclib was given at 300 mg daily for 21/28 days and everolimus was given continuously at 2.5 mg daily in 28 day cycles. The primary endpoint was progression free rate at 16 weeks. A Simon two-stage design was utilized and if at least 8 out of 24 pts were progression free at 16 weeks, the treatment was declared promising for the cohort. Here in we present data on the DDL cohort. Results: To date, 21 DDS pts, median age of 63 (range 40-79), of which 43% (n = 9) female were treated. Median prior lines of therapy was 1 (range 0-6). Of 19 pts with complete data, 8 (42%) met the primary endpoint of non-progression at 16 weeks. Confirmed partial response was seen in 2 pts (10%). Median PFS was 16 weeks, and stable disease occurring as best response in 11 (55%) pts. Grade 3-4 toxicities were uncommon except for lymphopenia (24%) and neutropenia (33%); no episode of neutropenic fever were observed. There was one death on study secondary to myocardial infarction, considered possibly related to therapy. Results of optional tissue biopsies pre and on therapy obtained to assess pharmacodynamic changes in PTEN, pAKT, CDK4, Rb and pS6 will be presented. Conclusions: The combination of ribociclib and everolimus demonstrates activity in DDL with prolonged stable disease (>16 weeks) meeting the primary protocol endpoint. Notably partial responses were also observed. The combination was well tolerated with acceptable side effects. Updated outcomes will be presented. Clinical trial information: NCT03114527.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 4525-4525 ◽  
Author(s):  
Noah M. Hahn ◽  
Thomas Powles ◽  
Christophe Massard ◽  
Hendrik-Tobias Arkenau ◽  
Terence W. Friedlander ◽  
...  

4525 Background: Anti-PD-L1 immunotherapy shows promising clinical activity in UC. We report a planned update of the safety and efficacy of durvalumab in patients (pts) with locally advanced/metastatic UC from a multicenter, phase 1/2 open-label study. Methods: Pts received durvalumab 10 mg/kg every 2 weeks (Q2W) up to 12 months (mo) or until unacceptable toxicity, progression, or starting another anticancer therapy. Primary endpoints were safety and confirmed objective response rate (ORR) by blinded independent central review (RECIST v1.1). Duration of response (DoR), progression-free survival (PFS) and overall survival (OS) were key secondary endpoints. Tumor PD-L1 expression was assessed by Ventana SP263 assay (PD-L1 high = ≥25% PD-L1 expression on tumor or immune cells). Results: As of Oct 24, 2016 (data cutoff [DCO]), 191 pts had received treatment. Median follow-up was 5.78 mo (range, 0.4–25.9). All pts had Stage 4 disease and 99.5% had prior anticancer therapy (95.3% post-platinum). As of DCO, ORR was 17.8% (34/191), including 7 CRs, with responses observed regardless of PD-L1 status (Table). Responses occurred early (median time to response, 1.41 mo) and were durable (median DoR not reached [NR]). Median PFS and OS were 1.5 mo (95% CI, 1.4, 1.9) and 18.2 mo (95% CI, 8.1, not estimable [NE]), respectively; the 1-year OS rate was 55.0% (95% CI, 43.9%, 64.7%). Grade 3/4 treatment-related AEs occurred in 6.8% of pts; grade 3/4 immune-mediated (im)AEs occurred in 4 pts; 2 pts discontinued due to imAEs (acute kidney injury and autoimmune hepatitis). Conclusions: Durvalumab 10 mg/kg Q2W shows favorable clinical activity and an excellent safety profile in locally advanced/metastatic UC pts. Table. Antitumor activity in UC pts, including second-line or greater (≥2L) post-platinum pts Clinical trial information: NCT01693562. [Table: see text]


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 871-871 ◽  
Author(s):  
Donna Weber ◽  
Ashraf Z. Badros ◽  
Sundar Jagannath ◽  
David Siegel ◽  
Victoria Richon ◽  
...  

Abstract Bortezomib, a proteasome inhibitor, is extremely effective for the treatment of multiple myeloma (MM). However, nearly all patients will eventually become refractory to bortezomib. Vorinostat is a potent inhibitor of histone deacetylase enzymes, and has been shown to affect growth of various cell lines, including MM, in a variety of in vitro non-clinical studies. Furthermore, the combination of bortezomib and vorinostat has demonstrated synergy in several in vitro and murine models. Subsequently, 2 multicenter, open-label, Phase I clinical trials have been conducted to investigate the combination of vorinostat with bortezomib in patients with MM. In the first trial, 34 patients with relapsed/refractory MM were enrolled. Patients received escalating doses of vorinostat (200 mg bid or 300–400 mg daily for 14 days) and bortezomib (0.7, 0.9, 1.1, or 1.3 mg/m2 on days 1, 4, 8, and 11); cycles were repeated every 21 days for ≤8 cycles or until progressive disease (PD) or intolerable toxicity. In the event of PD, oral dexamethasone (20 mg on days 1–4 and 17–20) could be added to the bortezomib plus vorinostat combination. The highest dose level of vorinostat was 400 mg daily for 14 days and bortezomib 1.3 mg/m2. The maximum tolerated dose (MTD) was not determined because ≥2 dose-limiting toxicities (DLTs) did not occur at any dose level. The most common drug-related adverse events were nausea (61.8%), diarrhea (58.8%), thrombocytopenia (50%), and vomiting (50%). Two patients experienced a DLT; grade 3 transient AST elevation was experienced by 1 patient receiving 400 mg vorinostat daily and 0.9 mg/m2 bortezomib, and grade 4 thrombocytopenia was experienced by 1 patient receiving 400 mg vorinostat daily and 1.3 mg/m2 bortezomib. Among 34 evaluable patients, the best response to vorinostat plus bortezomib was a partial response (PR) in 9 (26%) patients, minimal response (MR) in 7 (21%) patients, and stable disease (SD) in 18 (53%) patients. Mean duration of SD was 89 days, range 9–369 days. Of the 13 evaluable patients who had previously been treated with bortezomib, 5 achieved a PR, 1 had a MR, and 7 had SD. The effect of adding dexamethasone will also be analyzed and presented. The second trial enrolled 23 patients who received vorinostat (100–500 mg on days 4–11) and bortezomib (1–1.3 mg/m2 on days 1, 4, 8, and 11). Dexamethasone was added at cycle 2 for 6 patients who achieved less than a PR, and at cycle 4 for 5 patients with PD and at cycle 6 for 2 patients with PD. There was no upgrade in response for any patients who received additional dexamethasone. Two patients in the vorinostat 500 mg group experienced DLT (fatigue and prolonged QTc); MTD was identified as vorinostat 400 mg plus bortezomib 1.3 mg/m2. The main toxicities in this trial were hematologic (anemia, neutropenia, and thrombocytopenia). Twenty-one patients were evaluable for response (2 achieved very good PR, 7 PR, 10 SD, 2 progressive PD). Of the 9 patients who were refractory to bortezomib, 3 had PR, 4 had SD, 1 had PD, and 1 was non-evaluable. Co-administration of bortezomib did not alter the pharmacokinetics of vorinostat. In conclusion, these data suggest that the combination of vorinostat plus bortezomib is active for treatment of MM, even among some patients with prior exposure to bortezomib.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 888-888
Author(s):  
Bijal D. Shah ◽  
Wendy Stock ◽  
William G Wierda ◽  
Olalekan Oluwole ◽  
Houston Holmes ◽  
...  

Abstract Background : Approximately 45% of new ALL cases occur in adults ≥ 20 years of age (Howlader et al. SEER Cancer Statistics. 2015), and approximately 50% of adult patients relapse with poor subsequent outcomes (Oriol et al. Haematologica. 2010; Basson et al. JCO. 2011). Promising early efficacy and manageable safety were previously reported with anti-CD19 CAR T cells (KTE-C19) in adult patients with R/R ALL (Shah et al. ASCO 2017. #3024). Here we report updated results of the ZUMA-3 trial. Methods : Adult patients (≥ 18 years of age) with R/R ALL (Philadelphia+ eligible), > 5% bone marrow (BM) lymphoblasts; Eastern Cooperative Oncology Group performance status (ECOG) 0-1; and adequate renal, hepatic, and cardiac function were eligible. Patients with active graft-versus-host disease or clinically significant infection were not eligible. Patients received a target dose of 1 × 106 CAR T cells/kg or 2 × 106 CAR T cells/kg after lymphodepletion with 25 mg/m2/day fludarabine for 3 days and 900 mg/m2/day cyclophosphamide given on the last day. The primary endpoint of phase 1 was incidence of dose-limiting toxicities (DLTs). Key secondary endpoints included incidence of adverse events (AEs), incidence of minimal residual disease-negative (MRD-) responses, duration of remission (DOR), relapse-free survival (RFS), and overall survival (OS). Exploratory endpoints included levels of anti-CD19 CAR T cells in blood and levels of cytokines in serum. Results : As of the data cut-off date (DCO; April 26, 2017), 22 patients have been enrolled, and 16 patients received KTE-C19 on study. Four patients had not received treatment by the DCO, 1 patient did not receive KTE-C19 due to an AE after conditioning, and 1 patient received KTE-C19 under compassionate use. All 16 patients who received KTE-C19 prior to the DCO were included in the safety analysis, and all patients who had the opportunity to be followed for 8 weeks prior to the DCO were included in the efficacy analysis (n = 11). Of the 16 patients dosed with KTE-C19, 63% were male, 56% had ECOG 1, and 50% had received ≥ 2 previous lines of treatment, including 3 patients with prior blinatumomab. Nineteen percent of patients had undergone prior allogeneic stem cell transplant, 31% had R/R to ≥ second-line therapy, 31% had primary refractory disease, and 19% experienced first relapse within 12 months of first remission. Most patients (81%) had baseline BM blasts ≥ 60%. Six patients received the 2 × 106 cells/kg dose and 10 received the 1 × 106 cells/kg dose. No DLTs were observed. One patient experienced a grade 5 event of cytokine release syndrome (CRS) at the 2 × 106 cells/kg dose, and no other KTE-C19-related grade 5 AEs were observed. In the 16 patients who received KTE-C19, all of whom were followed for at least 4 weeks, the most common grade ≥ 3 AEs were hypotension (56%), anemia (50%), pyrexia (50%), and decreased platelet counts (44%). Grade ≥ 3 CRS and neurologic events (NE) were reported in 25% and 63% of patients, respectively. Tocilizumab (toci) or steroids were given for AE management in 94% and 75% of patients, respectively. In the 11 patients eligible for the efficacy analysis, objective response rate was 82%, including 8 (73%) patients with a complete remission (CR or CR with partial hematopoietic recovery), and 1 (9%) with blast-free BM. All remissions were MRD- as determined by flow cytometry. All 5 (100%) of the other patients who were too early for inclusion in the efficacy analysis had MRD- bone marrow with varying degrees of count recovery at the time of the DCO. Median follow-up was 6.8 months; 4 patients relapsed 63 - 168 days after treatment with KTE-C19. Efficacy was comparable between patients who recieved KTE-C19 doses of 1 × 106 and 2 × 106 CAR T cells/kg. Data from additional patients, including those treated with a lower dose of 0.5 × 106 CAR T cells/kg, as well as updated safety, efficacy, biomarker, and product characteristic analyses across dosing groups will be presented. Conclusions : In this ongoing phase 1 study, KTE-C19 has shown promising efficacy in adult patients with R/R ALL. The safety profile was generally manageable and additional approaches to improve the benefit:risk profile are being explored. ZUMA-3 continues to enroll additional patients at the 0.5 × 106 CAR T cells/kg dose level. Disclosures Wierda: AbbVie: Consultancy, Honoraria, Research Funding; Karyopharm: Research Funding; Genentech/Roche: Consultancy, Honoraria, Research Funding; Merck: Consultancy, Honoraria; Juno: Research Funding; Pharmacyclics: Consultancy, Honoraria, Research Funding; Gilead: Consultancy, Honoraria, Research Funding; Sanofi: Consultancy, Honoraria; Genzyme: Consultancy, Honoraria; Kite: Research Funding; GSK/Novartis: Consultancy, Honoraria, Research Funding; Emergent: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria; Janssen: Research Funding; The University of Texas MD Anderson Cancer Center: Employment; Acerta: Research Funding. Oluwole: Kite Pharma: Membership on an entity's Board of Directors or advisory committees. Schiller: Kite Pharma: Research Funding. Topp: Regeneron: Consultancy, Honoraria, Research Funding; Roche: Consultancy, Research Funding; Celgene: Other: Travel; Macrogenics: Consultancy, Research Funding; Amgen: Consultancy, Honoraria, Other: Travel, Research Funding. Kersten: Kite Pharma: Honoraria; Novartis: Honoraria; Roche: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Millenium/Takeda: Honoraria, Research Funding; Mundipharma: Honoraria; Gilead Sciences: Honoraria; BMS: Honoraria; MSD: Honoraria; Amgen: Honoraria. Mojadidi: Kite Pharma: Employment, Equity Ownership. Xue: Kite Pharma: Employment, Equity Ownership. Mardiros: Kite Pharma: Employment, Equity Ownership. Jiang: Kite Pharma: Employment, Equity Ownership. Shen: Kite Pharma: Employment, Equity Ownership. Aycock: Kite Pharma: Employment, Equity Ownership. Stout: Kite Pharma: Employment, Equity Ownership. Wiezorek: Kite Pharma: Employment, Equity Ownership. Jain: Kite Pharma: Employment, Equity Ownership.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 2592-2592
Author(s):  
Anthony B. El-Khoueiry ◽  
Jacob Stephen Thomas ◽  
Anthony J. Olszanski ◽  
Nilofer Saba Azad ◽  
Giles Francis Whalen ◽  
...  

2592 Background: Study IT-01 (KEYNOTE-A10) evaluates INT230-6, a novel formulation of cisplatin (CIS) and vinblastine (VIN) with an amphiphilic cell penetration enhancer designed for intratumoral (IT) administration, alone or in combination with pembrolizumab (PEM), an antibody to PD-1. INT230-6 dosing is set by a tumor’s volume. In preclinical studies, INT230-6 increases drug dispersion throughout the tumor, allows drug diffusion into cancer cells and recruits dendritic, CD4 and CD8 T cells. The addition of PEM has been shown to improve these responses in models. Phase 1 data indicated INT230-6 alone induced tumor regression in both injected and non-injected lesions. Considering the large volume of drug injected and retained in the tumor, coupled with immune infiltration on biopsies, RECIST response methodology may not capture the benefit of INT230-6 treatment. Methods: IT-01 is an open-label phase 1/2 study, currently enrolling adult subjects with solid tumors in phase 2. INT230-6 was administered IT Q2W for 5 doses alone or with PEM 200mg Q3W. The study seeks to assess the safety and efficacy of IT INT230-6 alone and in combination with PEM. Results: 67 subjects have been enrolled (58 mono and 12 INT230-6 + PEM (3 started in mono, then received combo)) having a median of 3 prior therapies (0, 10). Median age was 60 (42, 85). 20+ cancer types were accrued; breast cancer and sarcoma were the most frequent. Over 500 image guided INT230-6 IT injections were given (253 to deep tumors) at doses of 0.3 to 172mL (86 mg CIS, 17.2 mg VIN) in a single session, which are higher amounts than typical IV doses. PK shows that 95% of INT230-6 active agents remain in the tumor. The most common (> 20%) related TEAEs for INT230-6 alone were localized pain (57%), nausea (36%), fatigue (29%) and vomiting (24%); with grade 3 TEAEs (> 1) of localized pain (5%) and anemia (3%). The safety in the combination was similar. There were no related grade 4 or 5 TEAEs. In evaluable monotherapy subjects (n = 43), the disease control rate (DCR) was 65% vs. 100% in PEM subjects (n = 5). Given the range of dose and entering tumor burden, an exploratory analysis of dose relative to tumor burden (TB) showed that subjects receiving a dose of INT230-6 < 50% of their reported TB (n = 30) had a mOS of 3.5 months. While in subjects receiving a dose of INT230-6 to ≥50% of TB (n = 37), mOS has not yet been reached after a median follow up of 9.5 months (HR: 0.26 (0.13,0.51)). Conclusions: INT230-6 is well tolerated when administered IT as monotherapy and combined with PEM. Given the challenge in assessing overall response rate following IT delivery, an exploratory analysis suggests prolonged survival for subjects receiving an INT230-6 dose ≥50% of their tumor burden compares favorably to the < 50% group and to literature accounting for prognostic factors (ECOG, LDH, # of metastatic sites). Clinical trial information: 03058289.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1871-1871 ◽  
Author(s):  
Ashraf Z. Badros ◽  
Paul G. Richardson ◽  
Maher Albitar ◽  
Sundar Jagannath ◽  
Stefano Tarantolo ◽  
...  

Abstract Abstract 1871 Poster Board I-896 Introduction: Tanespimycin is an inhibitor of Hsp90, a molecular chaperone for proteins critical to growth, survival, and drug resistance of multiple myeloma (MM) cells. Increased Hsp70 expression is a pharmacodynamic (PD) marker of Hsp90 inhibition (Modi et al, J Clin Oncol, 2007). In primary MM cells, tanespimycin synergizes with bortezomib resulting in increased antitumor activity and enhanced proteasome inhibition. Tanespimycin + bortezomib has demonstrated durable responses in patients with relapsed/refractory MM with low rates of neutropenia and peripheral neuropathy (PN) relative to historical data on bortezomib monotherapy. Here we present the clinical and PD results of a randomized phase 2 study of tanespimycin + bortezomib in patients with relapsed/refractory MM that was truncated early for reasons unrelated to safety and efficacy. Methods: Patients with MM aged ≥18 years with Karnofsky performance status ≥70% and measurable disease who relapsed or progressed following ≥3 prior treatments (must have included lenalidomide and bortezomib) were eligible. Patients were randomized to 1 of 3 treatment arms with bortezomib 1.3 mg/m2 included in each arm: (1) tanespimycin 340 mg/m2, (2) tanespimycin 175 mg/m2, and (3) tanespimycin 50 mg/m2. Tanespimycin + bortezomib was administered on days 1, 4, 8, and 11 of each 21-day cycle. Treatment continued for at least 4 cycles and then until progression. Plasma Hsp70 levels and 20S proteasome activity as determined by chymotrypsin-like and caspase-like activity, were measured on days 1 and 11 at 0 and 4 hours postdose. Results: Twenty-two patients were randomized: 8 patients each in arm 1 and arm 2, and 6 patients in arm 3. Patients had a median age of 63 years; 64% were men, 46% had IgG subtype, 18% had IgA subtype, and 64% had heavy chain disease. At baseline, 3 patients had chromosome 13 deletion, 1 patient had t(11;14), 1 patient had t(14;16), 2 patients had other translocations, and 1 patient had p53 mutation. The median time since diagnosis was 81 months and the median number of prior regimens was 5 (range 3–12; 50% had received 5 or more prior regimens) and 96% had received prior thalidomide. A total of 72% of patients had had a prior stem cell transplant and 18% had not been candidates for transplant. For 68% of patients, the best response to the bortezomib-containing regimen prior to randomization had been stable disease or progressive disease. In the present study with tanespimycin + bortezomib, the overall response rate (≥ minimal response [MR]) was 14% including: 1 MR at 340 mg/m2 dose, and 1 very good partial response and 1 partial response (PR) at 175 mg/m2 dose. In addition, 10 patients had a best response of stable disease. The patient who achieved a PR had had 10 prior regimens including 3 bortezomib-containing regimens and had been refractory to the most recent regimen that included bortezomib + vorinostat. The most common adverse events (AEs) (any grade) were fatigue 73%, nausea 68%, diarrhea 64%, constipation 50%, and vomiting 46%; most AEs were Grade 1 or 2. PN was observed in 27% of patients; only 1 patient had Grade 3 PN that occurred after the patient had discontinued tanespimycin and was receiving bortezomib alone. Neutropenia (Grade 3) occurred in 18% of patients. Thrombocytopenia (any grade) occurred in 32% of patients; Grade 3/4 in 27%. From day 1 hour 0 to day 11 hour 0, Hsp70 levels increased 3.2-fold, 5.0-fold, and 6.3-fold in the 50 (n=3), 175 (n=3), and 340 mg/m2 (n=4) arms, respectively. Plasma chymotrypsin activity per μg proteasome at day 1 hour 4 was 90%, 77%, and 61% of baseline (day 1 hour 0) for the 50, 175, and 340 mg/m2 arms, respectively; at day 11 hour 4, chymotrypsin activity was 93%, 33%, and 40% of baseline for the 3 doses, respectively. A similar trend was observed for caspase activity per μg proteasome. Conclusion: In this randomized study, tanespimycin + bortezomib treatment resulted in a low frequency of PN and neutropenia, similar to a prior phase 1/2 study in patients with relapsed/refractory myeloma. Tanespimycin + bortezomib had activity in heavily pretreated myeloma patients (median 5 prior regimens; median 81 months since diagnosis), including a PR in a patient refractory to prior bortezomib + vorinostat. PD analysis confirms that tanespimycin + bortezomib effectively targets Hsp90 and the proteasome. Disclosures: Richardson: Millennium and Celgene: Speakers Bureau; Millennium Pharmaceuticals, Inc.: Research Funding. Jagannath:Millennium and Merck: Membership on an entity's Board of Directors or advisory committees. Berman:Bristol-Myers Squibb: Employment. Anderson:Celgene, Novartis, Millennium, BMS : Honoraria; Celgene, Novartis, Millennium, BMS: Research Funding; Celgene, Novartis, Millennium, BMS: Consultancy.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 1049-1049
Author(s):  
Yongheng Liu ◽  
Wei Lian ◽  
Xi Zhao ◽  
Wei Qi ◽  
Jian Xu ◽  
...  

1049 Background: HER2 is an effective therapeutic target for breast and gastric cancer. A166 is an antibody-drug conjugate composed of a novel cytotoxic drug site-specifically conjugated to transtuzumab sequence via a stable protease-cleavable valine citrulline linker. Methods: This was a single arm, open-label, multicenter, dose escalating Phase 1 first-in-human study of A166 as monotherapy in solid tumor patients. Dose escalation and MTD identification was directed using a Bayesian logistic regression model with overdose control. The following dose levels were evaluated in this study: 0.3, 1.2, 3.6, 4.8 mg/kg. (ClinicalTrials.gov NCT03602079) Results: As of November 1, 2019 35 pts have completed the DLT evaluation period across 4 dose levels. Overall, A166 had an acceptable toxicity profile with no unexpected toxicities related to the study drug. No adverse events recorded met the protocol specified definition of a dose limiting toxicity at any studied dose level. Most frequently (≥10%) occurring TEAEs include were Keratitis, Decreased appetite, Dry eye, Vision blurred etc. Overall incidence of ophthalmic toxicities in the 3.6 mg/kg cohort was 80% and in the 4.8 mg/kg cohort it was 83%. Among the 27 patients evaluable for efficacy, best response was progression of disease in 11 patients (41%), stable disease in 9 patients (33%) and partial response in 7 patients (26%), for the total disease control rate of 59%.Responses were seen only at the dose levels of 3.6 mg/kg and 4.8 mg/kg. Conclusions: A166 demonstrated clinically meaningful efficacy in heavily pretreated patients with relapsed or refractory advanced solid cancers. The achievement of an ORR of 36% at efficacious dose levels and up to 100% in HER2 positive patients regardless of histology (2 CRC, 1 BC and 1 NSCLC) at the highest studied dose level exceed Clinical trial information: NCT03602079 .


2019 ◽  
Vol 5 (suppl) ◽  
pp. 93-93
Author(s):  
Howard A. Burris III ◽  
Manish R. Patel ◽  
Daniel C. Cho ◽  
Jeffrey Melson Clarke ◽  
Martin Gutierrez ◽  
...  

93 Background: T-cell targeting of mutation-derived epitopes (neoantigens) has been demonstrated to drive anti-tumor responses. Immunizing patients against such neoantigens in combination with a checkpoint inhibitor (CPI) may elicit greater anti-tumor responses than CPI alone. Mutations are rarely shared between patients, thus requiring a personalized approach to vaccine design. Methods: This is an interim report of a phase I dose escalation study of mRNA-4157 given as monotherapy in patients with resected solid tumors and in combination with pembrolizumab in patients with unresectable solid tumors. mRNA-4157 is a lipid encapsulated personalized vaccine encoding multiple neoantigens selected using a proprietary algorithm designed to induce neoantigen specific T cells and associated anti-tumor responses. Patients received up to 9 cycles (Q3W) of mRNA-4157 by IM injection (0.04 – 1 mg). In combination arm, pembrolizumab (200 mg) was administered for two cycles prior to combination with mRNA-4157 for up to 9 cycles and may continue on pembrolizumab monotherapy for up to 2 years. Results: As of 10-May-2019, 33 patients received mRNA-4157 alone or in combination. No DLTs or related SAEs or AEs ≥ grade 3 were reported. Of the 13 patients treated with monotherapy (3 melanoma, 8 NSCLC, 2 MSI-high CRC), 11 patients remain disease free on study, median follow-up of 10 months. Of the 20 patients treated in combination (1 TMB-high metastatic cutaneous squamous cell, 4 bladder, 2 HNSCC, 1 melanoma, 7 NSCLC, 2 SCLC, 3 MSI-high (CRC, prostate, endometrial), 13 had received prior CPI, 5 PRs (2 in patients previously treated with PD-1/L1 inhibitors), 6 SD, and 8 PD were reported. Neoantigen specific CD8+ T-cell responses have been detected. Conclusions: mRNA-4157 is safe and well tolerated at all dose levels tested. Clinical responses have been observed in combination with pembrolizumab and neoantigen-specific T cells have been induced, supporting the advancement of mRNA-4157 to phase 2. Clinical trial information: NCT 03739931.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 4515-4515 ◽  
Author(s):  
Primo Lara ◽  
Todd Michael Bauer ◽  
Omid Hamid ◽  
David C. Smith ◽  
Thomas Gajewski ◽  
...  

4515 Background: Epacadostat (E) is a potent oral inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1), a tryptophan-catabolizing enzyme that induces immune tolerance by T-cell suppression. Preclinical and clinical data suggest that epacadostat has antitumor activity when combined with checkpoint inhibitors, including the PD-1 inhibitor pembrolizumab (P). ECHO-202/KEYNOTE-037 is an ongoing open-label, phase 1/2 (P1/2) study evaluating E + P in multiple tumor types. We report preliminary P1/2 efficacy and safety data for the advanced renal cell carcinoma (RCC) cohort as of a 29OCT2016 data cutoff. Methods: Eligible patients (pts) had advanced clear-cell RCC, prior antiangiogenic therapy (tx), and no prior checkpoint inhibitor tx. In P1 dose escalation (3+3+3), pts received E (25, 50, 100, or 300 mg PO BID) + P (2 mg/kg or 200 mg IV Q3W); MTD was not exceeded. E (100 mg BID) + P (200 mg Q3W) dosing was selected for P2 cohort expansion. Response was assessed in RECIST 1.1 evaluable pts. Safety/tolerability was assessed in pts receiving ≥1 E + P dose. Results: 33 pts (P1, n = 11; P2, n = 22) were enrolled (median age, 63 years; 70% men; 97% white; MSKCC criteria of favorable, intermediate, and poor in 6%, 64%, and 12% of pts, respectively). Of 30 efficacy-evaluable pts, 63% (n = 19) had 0–1 prior tx and 37% (n = 11) had ≥2 prior tx for advanced disease. ORR (CR+PR) and DCR (CR+PR+SD) for pts with 0–1 prior tx was 47% (9/19; 1 CR, 8 PR) and 58% (11/19; 1 CR, 8 PR, 2 SD), respectively; for pts with ≥2 prior tx, ORR and DCR were 0% and 36% (4/11; all SD). At data cutoff, 9/9 responses were ongoing (range, 1+ to 372+ days). PFS and biomarker analyses are ongoing. TRAEs occurring in ≥10% of the 33 pts included fatigue and rash (36% each); and arthralgia, diarrhea, pruritus, and pyrexia (12% each). Grade ≥3 TRAEs occurred in 15% of pts (none in > 1 pt). Two pts discontinued due to TRAEs (grade 3 autoimmune hepatitis, n = 1; grade 3 aseptic meningitis/headache/nausea/vomiting/anxiety, n = 1). Conclusions: E + P was generally well tolerated and associated with encouraging response outcomes in advanced RCC pts with 0–1 prior line of tx. E + P represents a novel immunotherapeutic strategy. A phase 3 RCC study is planned. Clinical trial information: NCT02178722.


Sign in / Sign up

Export Citation Format

Share Document