scholarly journals 482 Phase1/2 study of an anti-galectin-9 antibody, LYT-200, in patients with metastatic solid tumors

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A512-A512
Author(s):  
Aleksandra Filipovic ◽  
Zev Wainber ◽  
Judy Wang ◽  
Johanna Bendell ◽  
Filip Janku ◽  
...  

BackgroundGalectin-9 (gal-9) acts as a pivotal immuno-suppressor that disables immune mediated activity through modulation of T cells, macrophages and other immune functions. As such it has emerged as a powerful biological target for cancer immunotherapy and a potential biomarker of response and/or prognosis. Patients exhibiting high gal-9 expression in tumors and blood often have poor prognosis and tumors with aggressive and immunosuppressed molecular features (Chen L. et al, AACR 2020-LB-350). LYT-200 is a fully human IgG4 monoclonal antibody targeting gal-9. LYT-200 has high affinity, high specificity, stability, and blocks galectin-9 interactions with its binding partners in biochemical and human cell-based assays. In murine models of melanoma and pancreatic cancer, LYT-200 significantly reduced tumor growth, extended survival and modulated the intra-tumoral immune microenvironment. LYT-200 treated patient derived tumor organoids showed an increase in T cell activation (Chen L. et al, SITC 2019-P765).MethodsLYT-200 is now being evaluated in the USA, in the first part of an adaptive Phase 1/2 trial (NCT04666688) in relapsed/refractory solid tumors. Patients with solid tumor malignancy that is metastatic or unresectable and refractory to prior therapy are included. Patients are treated with LYT-200 by IV infusion, every 2 weeks (Q2W), until disease progression or toxicity. Phase 1 of the study uses the continuous reassessment design (CRM), and entails recruiting two patients per dosing level. Starting dose level was 0.2mg/kg Q2W. Additionally, the protocol stipulates six patients must be treated at the dose level intended to be declared recommended phase 2 dose (RP2D), for more robust assessment of safety/tolerability. RP2D may be the maximum tolerated dose or the optimal biological dose. The primary objective of the ongoing Phase 1 is to assess the safety and tolerability of LYT-200 and to identify the RP2D. The Phase 1 is also assessing LYT-200’s pharmacokinetics, immunogenicity and pharmacodynamics (measuring circulating gal-9 and cytokine levels, immunophenotyping peripheral blood mononuclear cells and tumor tissue). Preliminary efficacy is captured as an exploratory endpoint in Phase 1. Phase 2 expansion cohorts would implement the Simon’s two-stage design to further assess LYT-200 as a single agent and/or in combination with chemotherapy and tislelizumab. Phase 2 is currently planned in pancreatic cancer and other/different tumor types for Phase 2 may be guided by results of the Phase 1.AcknowledgementsAll clinical trial sites participating in the LYT-200 study. Shohei Koide, Linxiao Chen and George Miller and their teams at New York University Langone Health & New York University School of Medicine, NY for all the preclinical work on LYT-200.Trial RegistrationNCT04666688

2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 3013-3013 ◽  
Author(s):  
Michael Friedlander ◽  
Tarek Meniawy ◽  
Ben Markman ◽  
Linda R. Mileshkin ◽  
Paul R. Harnett ◽  
...  

3013 Background: The release of tumor-associated antigens may enhance the response to immunotherapy. BGB-A317, a humanized IgG4 variant monoclonal antibody engineered to have no Fc gamma receptor binding, targets the programmed cell death-1 (PD-1) receptor. It is being developed in solid and hematologic malignancies at a dose of 200 mg IV Q3W. BGB-290, a potent inhibitor of PARP 1/2, is hypothesized to promote neoantigen release that will potentially increase the efficacy of BGB-A317. A phase 1 study identified 60mg BID as the recommended Phase 2 dose (RP2D) for BGB-290. This study consists of initial dose escalation to determine the maximum-tolerated dose (MTD), safety, PK profile, and preliminary anti-tumor activity of the combination, followed by expansion into ovarian, breast, prostate, gastric, bladder, pancreatic and small cell lung cancers. Methods: Cohorts of 6 -12 pts with advanced solid tumors were treated in each of 5 planned dose levels (DLs). In DLs 1-3, BGB-290 doses ranged between 20-60mg PO BID with BGB-A317 2mg/kg IV Q3W. In DLs 4 - 5, BGB-290 doses were 40 or 60 mg BID; A317 was given at 200 mg IV Q3W based on PK data from a single agent Phase 1 study. Results: As of 16 Jan 2017, 38 pts [median age 59 years (34-75)] were treated in DLs 1-4; enrollment to DL5 is ongoing. One DLT of persistent Gr 2 nausea was reported in DL 4. The most common adverse event (AE) considered related to both study drugs was fatigue (10.5%). Immune-related AEs were Gr 3 hypophysitis (n = 1), Gr 3 or 4 autoimmune hepatitis(n = 2), and Gr 2 elevated AST/ALT (n = 1). Decreases in tumor burden have been observed in 16 pts; 7 achieved a PR (5 with ovarian and one each with uterine and pancreatic cancer) and one CR was observed in ovarian cancer. Six pts had SD for > 6 months including 2 pts with pancreatic cancer who received BGB-A317+BGB-290 for 189 and 281 days. Plasma/serum exposure of BGB-290 and BGB-A317 were consistent with those in single-agent trials. Conclusions: BGB290 and BGB-A317 can be combined. Dose expansion in multiple tumor types is planned to commence in 2017 once the RP2D is determined. Clinical trial information: NCT02660034.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. TPS2671-TPS2671
Author(s):  
Mark R. Middleton ◽  
Joseph J. Sacco ◽  
Jaime R. Merchan ◽  
Brendan D. Curti ◽  
Ari M. Vanderwalde ◽  
...  

TPS2671 Background: RP1 is an attenuated oncolytic HSV-1 that expresses a fusogenic glycoprotein from gibbon ape leukemia virus (GALV-GP R-) and GM-CSF. RP1 induces potent GALV-GP R- enhanced immunogenic cell death and host anti-tumor immunity in murine tumor models and increases PD-L1 expression. This clinical trial (NCT03767348) was designed to test the hypotheses that RP1 is safe when given alone and together with nivolumab (phase 1) and has efficacy together with nivolumab in four tumor types (phase 2). Methods: The primary goals of this clinical trial in a total of ~150 patients are to define the safety profile of RP1 alone and together with nivolumab, determine the recommended phase 2 dose (phase 1), and then in four phase 2 cohorts, to determine objective response rate in patients with melanoma, non-melanoma skin cancer, urothelial carcinoma and MSI-H solid tumors. Secondary objectives include duration of response, CR rate, PFS, viral shedding, and immune biomarker analysis. Patients with advanced cancer who failed prior therapy were eligible for the phase I component. In Phase 2 patients with histologic diagnoses of the four tumor types (N=30 for each) and who meet safety criteria for nivolumab treatment are eligible. Prior treatment with checkpoint blockade is not allowed except for the melanoma cohort. In the phase 1 portion patients are treated by intra-patient dose escalation of virus (range, 104 - 108 PFU) by intratumoral injection every two weeks for 5 total doses followed by 12 patients dosed 8 times at the RP2D in combination with nivolumab. Phase 1 patients were divided into two groups based on presence of clinically accessible lesions amenable to direct injection or those with visceral/deep lesions requiring image guidance for injection. In the phase 2 portion patients will receive the RP2D for eight injections and nivolumab will be given starting with the second RP1 injection. For the phase 1 portion, a modified 3+3 dose escalation design is used to assess safety and in the phase 2 portion, statistical analysis will be performed using a two-stage three-outcome optimum design with objective responses determined by RECIST criteria. As of February 11, 2019, 27 patients have been enrolled. Clinical trial information: NCT03767348.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 11057-11057 ◽  
Author(s):  
Erlinda Maria Gordon ◽  
Victoria S. Chua-Alcala ◽  
Katherine Kim ◽  
Rekha Baby ◽  
Nicole Angel ◽  
...  

11057 Background: Immune checkpoint inhibitors that promote sustained T cell activation may have synergistic activity with an mTOR inhibitor. This phase 1/2 study is aimed to investigate if ABI-009 a novel albumin-bound mTOR inhibitor is feasible and improve clinical outcomes in combination with nivolumab. Methods: Eligible patients with advanced UPS, LPS, CS, OS, or Ewing sarcoma are treated with the standard dose of nivolumab (240 mg given IV every 3 weeks, Day 1 of every 21-day Cycle). ABI-009 will be given IV on Days 8 and 15 of each cycle starting on Cycle 2 following the 2nd nivolumab dose. Phase 1 portion is a dose-finding study using the 3+3 design. The starting dose of ABI-009 is 56 mg/m2, and sequentially escalating doses are 75 and 100 mg/m2. The primary endpoint is to identify the maximum-tolerated dose (MTD) of ABI-009 + nivolumab, secondary endpoints include disease control rate, progression-free survival (PFS), and overall survival (OS). Exploratory endpoints include correlation of PFS and OS with PD-L1 and other biomarkers. The Phase 2 part of study will enroll 31 additional patients to further assess efficacy and safety at the MTD. Results: 9 patients were treated in Phase 1 (n = 3 each dose level); 5/9 patients had OS, 3/9 CS, and 1 had Ewing sarcoma. No dose-limiting toxicities (DLTs) were observed, the MTD was not reached, and 100 mg/m2 ABI-009 was designated as the recommended phase 2 dose. Safety analysis: At Dose 1: Grade 3 treatment-related adverse events (TRAEs) included hyper dyslipidemia (n = 1), and hyperglycemia (n = 1). At Dose 2: Grade 3 TRAEs included increased ALT (n = 1). At Dose 3: Grade 3 TRAEs included hypophosphatemia (n = 1). Seven of 9 patients have discontinued treatment: 5 patients due to PD, 2 with SD opted to stop treatment due to drug-related Grade 2 AEs (pruritus, acneiform rash, and 2 with SD are still on therapy at Dose 3. The median PFS at dose level 3 has not yet been reached. Conclusions: The MTD was not reached and Dose 3 (100 mg/m2) has been designated as the phase 2 dose of ABI- 009, combinable with nivolumab. Enrollment to phase 2 is ongoing. Clinical trial information: NCT03190174.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 3080-3080
Author(s):  
Nashat Y. Gabrail ◽  
Erika P. Hamilton ◽  
Anthony D. Elias ◽  
Mothaffar F. Rimawi ◽  
Chao Li ◽  
...  

3080 Background: ORIN1001 is a first-in-class small molecule with a novel, unique enzyme and mode of inhibition that selectively inhibits Inositol Requiring Enzyme 1α (IRE1) RNAse and blocks X-Box Binding Protein 1 (XBP1) activation in the endoplasmic reticulum (ER). IRE1α/XBP1 has been implicated in a host of pathologies, and molecules that modulate it are under intense investigation for the treatment of oncologic, metabolic, neurodegenerative and other diseases. ORIN1001 has demonstrated preclinical anti-tumor activity alone and in combination with standard of care across multiple animal models including breast, prostate, lung, liver, pancreatic, brain, colon, ovarian, esophageal, and hematologic cancers and is now undergoing first-in-human testing. Methods: A phase 1, open label, 3+3 dose escalation trial is testing ORIN1001 administered PO daily to patients (pts) with advanced solid tumors (single agent) or relapsed refractory breast cancer (in combination with Abraxane). The phase 1 dose escalation part of the trial evaluates the safety, tolerability, pharmacokinetics and preliminary efficacy of ORIN1001. After identification of the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) for the single agent, the dose expansion part of the trial will test ORIN1001 in combination with Abraxane. Results: As of Jan 25, 2021, 22 patients with advanced cancer have received ORIN1001 dosed at 100mg, 200mg or 300mg per day in 21-day continuous cycles with a median age of 61 (range 42-77). The pts had received a median of 4 prior line of treatments. Two DLTs were observed at 200 mg with thrombocytopenia and rash. MTD has not been reached. Common (>15%) treatment-emergent adverse events (TEAEs) included nausea, vomiting, rash, fatigue, and hypokalaemia. The vast majority of these events were Grade 1-2 in severity. Seven (32%) pts had at least 1 TRAE grade≥ 3, the most frequent of which were thrombocytopenia (N=3) and rash (N=3). Preliminary pharmacokinetic analysis showed ORIN1001 exposure to increase in a dose proportional manner. Mean t1/2 at steady state was 18 hrs. Thirteen pts were evaluated for preliminary efficacy. Best response, per RECIST 1.1, was stable disease (SD) in 8 pts while 5 pts had progressive disease (PD). For 2 ongoing patients with advanced liver or colorectal cancer, duration of treatment has exceeded 300 days and 570 days, respectively. Conclusions: To date, the phase 1 part of the first-in-human trial has demonstrated a reasonable safety and pharmacokinetic profile for ORIN1001 at 100mg and 200mg dose levels. While efficacy data have yet to mature, chronic dosing achieved in pts with heavily treated advanced solid tumors, suggests clinical potential for in the setting of advanced solid cancers. The phase 2 part of the trial testing ORIN1001 in combination with Abraxane is currently enrolling pts with advanced breast cancer. Clinical trial information: NCT03950570.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS3149-TPS3149
Author(s):  
Melissa Lynne Johnson ◽  
Deborah Blythe Doroshow ◽  
Tanguy Y. Seiwert ◽  
Michael K. Gibson ◽  
Vamsidhar Velcheti ◽  
...  

TPS3149 Background: Dependence of cancer cells on glutamine has made glutaminolysis an attractive therapeutic target in cancer. Prior clinical trials evaluating glutamine analogues for the treatment of cancer were abandoned due to lack of efficacy and/or tolerability. DON (6-Diazo-5-oxo-L-norleucine) is an irreversible inhibitor of several enzymes that utilize glutamine as a metabolic substrate. In addition to direct anti-tumor efficacy, inhibition of glutamine metabolism in the tumor microenvironment has been shown to improve T-cell activation and tumor infiltration, increasing anti-tumor immune responses. As such, combining DON with an immune checkpoint inhibitor (ICI), has strong preclinical rationale. The investigational product DRP-104 (sirpiglenastat) is an inactive prodrug of DON designed to limit systemic DON exposure while targeting glutamine dependence in tumor cells. Methods: A phase 1/2a, FIH, multi-center, non-randomized, multi-cohort, open-label study of DRP-104 is currently open to accrual for patients with advanced solid tumors. This study will be conducted in 4 parts: A) Dose Escalation of IV and subQ DRP-104 (Run-In phase followed by modified Continual Reassessment Method) to define MTD/RP2D. Primary objective of dose escalation is to assess the safety, tolerability, pharmacokinetics, pharmacodynamics and preliminary antitumor activity of DRP-104 as a single agent; B) Dose Expansion of IV and subQ DRP-104 for safety assessment while primary objective is to select and recommend phase 2 DRP-104 route of administration; C) Phase 2a at recommended MTD/RP2D of selected route of DRP-104 in 2 patient cohorts: patients with locally advanced/metastatic NSCLC with KEAP1, NFE2L2 and/or STK11 mutation and patients with unresectable or metastatic SCCHN, in order to assess the safety, tolerability and preliminary antitumor activity of DRP-104 as a single agent; D) Phase 2a at recommended MTD/RP2D of selected route of DRP-104 in combination with atezolizumab in adult patients with advanced solid tumors previously treated with an ICI, in order to assess the safety, tolerability and preliminary antitumor activity of DRP-104 in combination with atezolizumab; DRP-104 IV is infused TIW over 1 hour infusion for 2 consecutive weeks followed by 1 week off. DRP-104 subQ is administered BIW weekly. Study is currently open with 6 IV patients (Run-In Phase completed and at Dose Level 4) and 3 subQ patients at Dose Level 1 at time of submission. Clinical trial information: NCT04471415.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 3515-3515 ◽  
Author(s):  
P. J. Rosen ◽  
M. Gordon ◽  
P. N. Lee ◽  
E. Sausville ◽  
K. P. Papadopoulos ◽  
...  

3515 Background: CFZ is a novel proteasome inhibitor of the peptide epoxyketone class that exhibits a high level of selectivity for proteasome active sites. This phase 1/2 study assessed the maximum tolerated dose (MTD), safety, efficacy, pharmacokinetics (PK), and pharmacodynamics (PD) of CFZ in patients (pts) with advanced metastatic solid tumors. Methods: Pts failing ≥ 2 prior treatments were enrolled in the phase 1 3+3 dose escalation study. Pts received CFZ IV Day (D) 1, 2, 8, 9, 15 and 16 every 28 d for up to 12 cycles (C) Cycle 1 D1, D2 dosing in all cohorts was at 20 mg/m2. Subsequent doses remained at 20 mg/m2 or were escalated to 27 or 36 mg/m2 in a stepped up regimen on D8. At 20/36 mg/m2, 1 pt had a DLT (Grade 3 fatigue) and established the phase 2 dose. Phase 2 is designed as a Simon 2 stage of 70 pts split into 5 subgroups (small cell lung [SCLC], non-small cell lung [NSCLC], ovarian, renal, and other cancer). Tumor response was measured every 2 C. ORR, defined as CR+PR+SD, to 16 wks of CFZ. Stage 2 will open if a 1 PR or better at 16 wks occurs in a selected subgroup. Results: 14 pts in phase 1 and 51 pts in phase 2 (23M/28F, mean age 61 yrs) received a total of 154.5 cycles of CFZ. Median cycles administered was 1.7 (range 1 to 12). To date, there were 6 SCLC, 10 NSCLC, 11 ovarian, 6 renal, and 18 other cancer patients enrolled to the Simon stage 1. Efficacy of SD or better is detailed in the table. The most common AEs included fatigue headache, diarrhea, nausea and constipation. Notable was the absence of grade > 1 peripheral neuropathy and severe hematologic toxicities. Final results of the PK and PD will be reported. Conclusions: CFZ is active as a single agent in relapsed solid tumors demonstrating PR in both renal and SCLC; and SD >16 wks in mesothelioma, ovarian, renal and NSCLC. The 20/36 mg/m2 QDx2 dose schedule was well tolerated and lacks severe myelosuppression, hepatotoxicity and neuropathy which make CFZ an attractive agent to combine with traditional or novel targeted agents. [Table: see text] [Table: see text]


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 10019-10019
Author(s):  
Jennifer Foster ◽  
Joel M. Reid ◽  
Charles G. Minard ◽  
Emasenyie Isikwei ◽  
Xiaowei Liu ◽  
...  

10019 Background: Pevonedistat (PEV), a first in class inhibitor of NEDD8 activating enzyme (NAE), prevents the activation of Cullin-RING ligases (CRL) necessary for proteasome mediated degradation of key regulatory proteins important in cell survival. In adults with solid tumors, the maximum tolerated dose (MTD) in combination with chemotherapy is 20-25 mg/m2. Antitumor activity of PEV has been demonstrated in preclinical models of childhood cancer. In vivo additive activity has been demonstrated for PEV in combination with IRN and alkylating agents. The objectives of this study are to determine the MTD and recommended Phase 2 dose of PEV in combination with IRN and TMZ and describe the toxicities, pharmacokinetic (PK), and pharmacodynamics (PD) properties of this combination. Methods: We conducted a phase 1 trial of PEV in combination with IRN and TMZ in pediatric patients (pts) with recurrent or refractory solid tumors and brain tumors. During cycle 1, PEV was administered intravenously on days 1, 8, 10, and 12, with IRN (IV, 50mg/m2) and TMZ (orally, 100mg/m2), on days 8-12 of a 28 day cycle. In subsequent cycles, PEV was administered on days 1, 3, and 5, with IRN and TMZ on days 1-5 of a 21 day cycle. Dose escalation was determined using the Rolling 6 Design. Results: 30 pts enrolled. All pts were eligible and evaluable for cycle 1 dose limiting toxicity (DLT) assessment. Median (range) age was 13 (1-21) years; 19 (63%) were male. Eleven pts had brain tumors, and 19 pts had solid tumors. Six pts each enrolled on PEV dose levels (DL) 1 (15mg/m2), 2 (20mg/m2), 3 (25mg/m2) and 4 (35mg/m2) as well as an expanded PK cohort at DL4. Cycle 1 grade 3/4 toxicities include lymphopenia (n = 5), leukopenia (n = 4), neutropenia (n = 2), elevated ALT (n = 2), elevated AST (n = 1), diarrhea (n = 1), flu-like symptoms (n = 1). The most frequent non-dose limiting AEs in cycle 1 were anemia (87%), WBC decreased (77%), nausea (57%), diarrhea (53%), ALT increased (50%), AST increased (50%), and vomiting (50%). PK analyses showed the mean area under the curve at the 25 mg/m2 dose level on day 8 (in combination with irinotecan and temozolomide) was 1300 hr•ng/mL, half-life (T ½) was 5-6 hours, time to maximum concentration (Tmax) was 1 hour, and mean clearance was 20 L/hr/m2. There were 3 DLTs, 2 of which were related to protocol therapy (diarrhea and thrombocytopenia), among 12 patients on DL4. Thus the MTD was not exceeded at any dose level. PK at the 25 mg/m2 dose level are comparable to those in adult patients. PK from the 12 patients on DL4 (35mg/m2) as well as responses of all patients are pending. Conclusions: PEV in combination with IRN and TMZ is well tolerated in children with solid or brain tumors. PEV PK was not altered by the addition of irinotecan and temozolomide. Further PK and PD analyses are ongoing to establish the recommended phase 2 dose. Clinical trial information: NCT03323034.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi71-vi71
Author(s):  
Stefan Symeonides ◽  
Jeff Evans ◽  
Hendrik-Tobias Arkenau ◽  
Meredith McKean ◽  
Nehal Lakhani ◽  
...  

Abstract BACKGROUND C/EBPβ is a transcription factor that is active during embryofetal development but held in an inactive state in most mature cells (Zahnow 2009). C/EBPβ is upregulated or overactivated in multiple cancers, where it inversely correlates with disease prognosis and survival due to activation of a gene signature that promotes tumor cell proliferation and survival. ST101 is a cell-penetrating peptide antagonist of C/EBPβ. ST101 exposure leads to selective tumor cell death in multiple human cancer cell lines, including GBM, without impacting normal cell viability. In vivo, ST101 displays rapid uptake into multiple organs, the ability to cross the blood-brain barrier, and a long plasma half-life due to its resistance to degradations. It has potent anti-tumor activity in multiple GBM models, as a single agent or in combination, which supported moving into clinical development. TRIAL DESIGN This phase 1-2 study is enrolling patients ≥ 18 years of age with advanced, unresectable metastatic solid tumors refractory to or intolerant of other therapeutic options. We began recruitment in August 2020. The primary objective of phase 1 is to evaluate safety and tolerability of ST101. Secondary objectives include the recommendation of a dose and regimen of ST101 for further evaluation, pharmacokinetics, several pharmacodynamic measures, and preliminary efficacy. Patients receive intravenous ST101 once weekly in a standard 3 + 3 design. Enrollment is ongoing, and by 21 May 2021, 15 patients have been recruited in four dose-escalation cohorts up to 4 mg/kg; a 5th cohort (6 mg/kg) is ongoing. The recommended phase 2 dose will be used in a 15-30 patient GBM expansion cohort, with a Simon 2-stage design, which requires one response or two patients with PFS6 in the first cohort to continue the study. Up to 120 patients are planned in a total of four expansion cohorts, which should be enrolling by Q3 2021.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A852-A852
Author(s):  
Sumayah Jamal ◽  
Adnan Nagrial ◽  
Anthony Joshua ◽  
Richard Eek ◽  
Sumayah Jamal

BackgroundThe endothelin B receptor (ETBR) is upregulated in many types of cancer and is associated with poor overall survival and a paucity of TILs (tumor infiltrating lymphocytes). The ETBR prevents T-cell extravasation and tumor infiltration by a mechanism involving adhesion molecule downregulation in the tumor vasculature. Thus ETBR expression may mediate resistance to immunomodulatory therapy. ENB-003 is a small molecule ETBRi (ETBR inhibitor) which overcomes resistance to anti-PD1 across multiple cancer types in preclinical studies. Part 1 of this study seeks to evaluate the safety and tolerability of ENB-003 in combination with pembrolizumab in refractory advanced ETBR+ solid tumors. Part 2 of the study is an expansion cohort basket trial assessing the efficacy of ENB-003 in combination with pembrolizumab in anti-PD1 refractory melanoma, platinum resistant ovarian cancer and refractory pancreatic cancer.MethodsStudy ENB-003-101 (MK-3475-951) is a multicenter, Phase 1/2, open-label study of ENB-003 in combination with pembrolizumab in adult subjects with advanced solid tumors. The part 1 dose escalation is enrolling subjects with ETBR+ tumors and includes 5 doses of ENB-003 in combination with a fixed dose of pembrolizumab. The primary objective of part 1 is to assess safety and tolerability, the secondary objective is to evaluate anti-tumor effect (RECIST 1.1 and iRECIST). Exploratory objectives are to examine biomarkers/pharmacodynamics.ResultsENB-003, as a single agent and in combination with anti-PD1, was investigated in a variety of syngeneic preclinical models. ENB-003 enhanced the anti-tumor activity of anti-PD1 in anti-PD1 resistant models of melanoma, ovarian cancer, pancreatic cancer, bladder cancer and SCC. For example, the combination of ENB-003 plus anti-PD1 in an anti-PD1-resistant melanoma model resulted in complete tumor eradication in 21 days as well as the formation of TLOs (tertiary lymphoid organs). The combination of ENB-003 plus pembrolizumab was well tolerated in the first 2 cohorts of the ongoing Phase 1 trial in patients with advanced refractory solid tumors that are ETBR+. Best overall responses from the first 2 cohorts (n=6) demonstrates disease stabilization (SD) in 2 patients as well as a partial response (PR) in an ovarian cancer patient with ~60% reduction in target lesions.ConclusionsETBRi is a novel approach to overcoming immunotherapy resistance. The combination of ENB-003 and pembrolizumab is well tolerated thus far and is demonstrating promising early signals of anti-tumor efficacy. Trial updates will be reported.Trial RegistrationNCT04205227Ethics ApprovalThis study was approved by an institutional Review Board at each investigational site.ReferenceKandalaft L, Facciabene A, Buckanovich R, Coukos G. Endothelin B Receptor, a New Target in Cancer Immune Therapy. Clin Cancer Res 2009;15(14):4521-4528.


2021 ◽  
Vol 9 (7) ◽  
pp. e002446
Author(s):  
Rachel E Sanborn ◽  
Omid Hamid ◽  
Elisabeth GE de Vries ◽  
Patrick A Ott ◽  
Javier Garcia-Corbacho ◽  
...  

BackgroundProbody® therapeutics are antibody prodrugs designed to be activated by tumor-associated proteases. This conditional activation restricts antibody binding to the tumor microenvironment, thereby minimizing ‘off-tumor’ toxicity. Here, we report the phase 1 data from the first-in-human study of CX-072 (pacmilimab), a Probody immune checkpoint inhibitor directed against programmed death-ligand 1 (PD-L1), in combination with the anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) antibody ipilimumab.MethodsAdults (n=27) with advanced solid tumors (naive to PD-L1/programmed cell death protein 1 or CTLA-4 inhibitors) were enrolled in the phase 1 combination therapy dose-escalation portion of this multicenter, open-label, phase 1/2 study (NCT03013491). Dose-escalation pacmilimab/ipilimumab followed a standard 3+3 design and continued until the maximum tolerated dose (MTD) was determined. Pacmilimab+ipilimumab was administered intravenously every 3 weeks for four cycles, followed by pacmilimab administered every 2 weeks as monotherapy. The primary objective was identification of dose-limiting toxicities and determination of the MTD. Other endpoints included the rate of objective response (Response Evaluation Criteria In Solid Tumors v.1.1).ResultsTwenty-seven patients were enrolled in pacmilimab (mg/kg)+ipilimumab (mg/kg) dose-escalation cohorts: 0.3+3 (n=6); 1+3 (n=3); 3+3 (n=3); 10+3 (n=8); 10+6 (n=6); and 10+10 (n=1). Dose-limiting toxicities occurred in three patients, one at the 0.3+3 dose level (grade 3 dyspnea/pneumonitis) and two at the 10+6 dose level (grade 3 colitis, grade 3 increased aspartate aminotransferase). The MTD and recommended phase 2 dose was pacmilimab 10 mg/kg+ipilimumab 3 mg/kg administered every 3 weeks. Pacmilimab-related grade 3–4 adverse events (AEs) and grade 3–4 immune-related AEs were reported in nine (33%) and six (22%) patients, respectively. Three patients (11%) discontinued treatment because of AEs. The overall response rate was 19% (95% CI 6.3 to 38.1), with one complete (anal squamous cell carcinoma) and four partial responses (cancer of unknown primary, leiomyosarcoma, mesothelioma, testicular cancer). Responses lasted for >12 months in four patients.ConclusionsThe MTD and recommended phase 2 dose of pacmilimab (10 mg/kg)+ipilimumab (3 mg/kg) every 3 weeks is active and has a favorable tolerability profile.


Sign in / Sign up

Export Citation Format

Share Document