372 Single-agent anti-tumor activity in relapsed/refractory solid tumors: interim data from the phase 1 solid tumor trial of AMV564, a novel T-cell engager

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A397-A397
Author(s):  
Sarina Piha-Paul ◽  
Alexander Starodub ◽  
Raghad Karim ◽  
Michael Shafique ◽  
Gabriel Tinoco Suarez ◽  
...  

BackgroundOvercoming the immune-suppressive tumor environment induced by myeloid-derived suppressor cells (MDSC) is a major challenge in immune therapy. AMV564 is a potent conditional agonist that engages T cells to selectively deplete target cells such as MDSC while promoting T cell polarization and activation. Whereas CD33 plays an insignificant role in differentiated myeloid cells, CD33 signaling in immature myeloid cells promotes expansion of MDSC and production of immune-suppressive factors. Preferential binding of AMV564 to areas of high CD33 density enables selective targeting of MDSC. Ex vivo data1 as well as data from a clinical trial in acute myeloid leukemia (NCT03144245) demonstrate the ability of AMV564 to selectively deplete MDSC while sparing monocytes and neutrophils.2 3MethodsNCT04128423 is a multi-center Phase 1 study to determine the safety and tolerability, define the maximum-tolerated or pharmacologically active dose, and assess the preliminary efficacy of AMV564. In this 3+3 dose escalation study, patients with advanced solid tumors receive AMV564 once daily via subcutaneous (SC) injection on Days 1–5 and 8–12 of a 21-day cycle. Primary endpoints include incidence, nature and severity of adverse events (AEs). Secondary endpoints include assessment of pharmacokinetics and pharmacodynamics.ResultsAs of June 30, 2020, 11 patients have been dosed across 3 dose cohorts (15 mcg – 75 mcg). The tumor types enrolled were: colorectal (n=2), GE junction (n=2), pancreatic (n=2), squamous cell carcinoma (n=2), small intestine, ovarian, and endometrial cancer. AMV564 has been well tolerated with no dose-limiting toxicities. The most common treatment-related AEs were fever/pyrexia (Grade 1: n=3; Grade 2: n=8) and injection site reactions (Grade 1: n=1; Grade 2: n=9). Preliminary estimate of median plasma half-life for AMV564 after SC injection was >48 hours, with dose-related increases in peak plasma concentration (Cmax). Tumor responses were evaluable in 9 patients; 1 patient had not reached their first assessment and 1 patient was not efficacy evaluable due to a non-treatment-related AE resulting in study discontinuation. Single-agent activity has been observed including a complete response by RECISTv1.1 criteria in 1 patient with ovarian cancer refractory to all standard therapies and anti-PD-1 therapy, and stable disease in 4 additional patients.ConclusionsAMV564 has been well tolerated across multiple dose levels, with good plasma exposure and evidence of anti-tumor activity when administered subcutaneously. Single-agent anti-tumor activity was observed in an ovarian cancer patient.AcknowledgementsWe would like to thank the patients and their families for participating in this clinical trial.Trial RegistrationNCT04128423Ethics ApprovalThe study was approved by the Institutional Review Board at each center where the study is being conducted.ReferencesCheng P, Eksioglu E, Chen X, et al. Immunodepletion of MDSC By AMV564, a novel Tetravalent bispecific CD33/CD3 T cell engager restores immune homeostasis in MDS in Vitro. Blood. 2017; 130:51 (abstract).Eckard S, Gehrs L, Smith V, et al. AMV564, a novel bivalent, bispecific T-cell engager, targets myeloid-derived suppressor cells. SITC Annual Meeting; 2019 Nov 6-10. Oral Presentation O71.Westervelt P, Roboz G, Cortes J, et al. Safety and Clinical Activity of AMV564, a CD33/CD3 T-cell Engager, in Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML): Updated Results from the Phase 1 First-in-Human Trial. EHA Annual Congress; 2019 Jun 13-16. Abstract S877.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2779-2779
Author(s):  
Cesarina Giallongo ◽  
Nunziatina Parrinello ◽  
Daniele Tibullo ◽  
Piera La Cava ◽  
Alessandra Cupri ◽  
...  

Abstract Abstract 2779 Background: Tumor cells are able to develop immune evasion mechanisms which induce a state of immune tolerance and inactivate tumor-specific T cells. In this context, in some solid tumors it has been demonstrated that a subpopulation of myeloid cells, defined as “myeloid-derived suppressor cells” (MDSCs), plays an important role in inducing T cell tolerance by production of arginase that depletes microenvironment of arginine, an essential aminoacid for T cell function. Since chronic myeloid leukemia (CML) patients have high levels of immature myeloid cells it is of interest to investigate if these cells have MDSCs phenotype and activity. Aim: The aim of this study was to analyze MDSCs and investigate their involvement in T-cell anergy of CML patients. Methods: MDSCs were analyzed in peripheral blood (PB) of 13 CML patients (at diagnosis and during therapy) and healthy donors (HD; n=20) by cytofluorimetric analysis (CD14+DR- for monocytic MDSCs and CD11b+CD33+CD14-DR- for granulocytic MDSCs). Arginase 1 expression was assessed in PB of HD and CML patient using real time PCR. Purification of granulocytes, monocytes and lymphocytes from PB was performed by a positive magnetic separation kit (EasySep, STEMCELL Technologies). Arginase activity was measured in granulocyte lysates using a colorimetric test after enzymatic activation and arginine hydrolysis. To evaluate the activation of CD3+ T lymphocytes after incubation with phytoemagglutinin, we analyzed at 24, 48, 72 h the following markers: CD69+, CD71+, DR+. Microvesicles were isolated from CML serum at diagnosis (n=5) by sequential ultracentrifugation. Results: CML patients showed high levels of monocytic and granulocytic MDSCs at diagnosis in comparison to HD (63±8 and 83±12,2% respectively in CML vs 4,9±2,1 and 55,8±5,3% respectively in HD; p<0.001) while after 3–6 months of tyrosine kinase inhibitors (TKIs) therapy MDSC levels returned to normal values. Either in PB and in the purified granulocytes subpopulation, arginase1 expression showed a 30 fold increase in CML at diagnosis (CML vs HD: p<0.01) and decreased after therapy. We also evaluated arginase enzymatic activity in granulocytes and we found it increased in CML patients (n=4) compared to HD (n=5) (p<0.05). CML as well as HD T lymphocytes showed a normal activation in vitro which was significantly lost when they was incubated with CML serum (n=4). In addition, an increase of monocytic MDSCs in vitro was observed after incubation of HD monocytes with CML serum (39±6%; p<0.01) or microvescicles (9,2±1,2%; p<0.05) compared to control serum. Conclusions: Granulocytic and monocytic MDSCs are increased in CML patients at diagnosis and decrease during TKIs treatment. Their levels also correlates with Arginase 1 expression and enzymatic activity in granulocytes. CML serum as well as CML microvesicles increase the percentage of HD monocytic MDSCs. Moreover, CML serum leads to anergy of T lymphocytes, probably by Arginase 1 secretion. Disclosures: Off Label Use: Eltrombopag is a thrombopoietin receptor agonist indicated for the treatment of thrombocytopenia in patients with chronic immune (idiopathic) thrombocytopenic purpura (ITP).


2017 ◽  
Vol 35 (7_suppl) ◽  
pp. 126-126
Author(s):  
Jaspreet Singh Grewal ◽  
Numan Al-Rayyan ◽  
Jamaal Ritchie ◽  
Paxton Schowe ◽  
Cam Falkner ◽  
...  

126 Background: Myeloid derived suppressor cells (MDSCs) inhibit the expansion of tumor antigen-specific effector CD8+ T cells via different mechanisms including increased expression of arginase, transforming growth factor – β (TGF – β) and indoleamine 2,3-dioxygenase (IDO). Recently, MDSCs were found to over-express hypoxia inducible factor 1 alpha (HIF-1α) which is required for their differentiation. An essential transcriptional target of HIF-1α is 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) which synthesizes fructose 2,6-bisphosphate, an allosteric stimulator of glycolysis and of proliferation via stimulation of cyclin dependent kinase-1 (CDK1). We hypothesized that MDSCs might over-express PFKFB3 which in turn might be required for their function as T cell suppressors. Methods: We used monocytic MDSCs (M-MDSCs) induced by co-culture with A375 melanoma cells, M-MDSCs from metastatic melanoma patients, murine bone marrow MDSCs and splenic M-MDSCs from B16 F10 tumor bearing mice for our studies. T cell suppression assays were performed to analyze M-MDSC suppression and reversal following PFKFB3 blockade. Results: We found that M-MDSCs have increased PFKFB3 expression. We also found that PFK-158 administration in B16 (wild-type) melanoma-bearing mice results in a marked reduction in MDSCs and a simultaneous increase in CD8+ T cell infiltration in the tumors. We analyzed three advanced cancer patients for circulating MDSCs before and after PFK-158 administration as part of a multi-center phase 1 clinical trial. And, we found that the MDSCs were markedly reduced in each patient. In addition, we have generated data for MDSC suppressive activity following in vitro treatment with PFK-158 showing reversal of suppressive activity. Conclusions: Taken together, these data indicate that selective inhibition of PFKFB3 may be a novel approach to target MDSCs and combinations of PFKFB3 inhibitors with immunotherapies may be a rational strategy to promote durable immune-mediated remissions in cancer patients.


Blood ◽  
2017 ◽  
Vol 129 (24) ◽  
pp. 3165-3174 ◽  
Author(s):  
Ramiro Garzon ◽  
Michael Savona ◽  
Rachid Baz ◽  
Michael Andreeff ◽  
Nashat Gabrail ◽  
...  

Abstract Selinexor is a novel, first-in-class, selective inhibitor of nuclear export compound, which blocks exportin 1 (XPO1) function, leads to nuclear accumulation of tumor suppressor proteins, and induces cancer cell death. A phase 1 dose-escalation study was initiated to examine the safety and efficacy of selinexor in patients with advanced hematological malignancies. Ninety-five patients with relapsed or refractory acute myeloid leukemia (AML) were enrolled between January 2013 and June 2014 to receive 4, 8, or 10 doses of selinexor in a 21- or 28-day cycle. The most frequently reported adverse events (AEs) in patients with AML were grade 1 or 2 constitutional and gastrointestinal toxicities, which were generally manageable with supportive care. The only nonhematological grade 3/4 AE, occurring in &gt;5% of the patient population, was fatigue (14%). There were no reported dose-limiting toxicities or evidence of cumulative toxicity. The recommended phase 2 dose was established at 60 mg (∼35 mg/m2) given twice weekly in a 4-week cycle based on the totality of safety and efficacy data. Overall, 14% of the 81 evaluable patients achieved an objective response (OR) and 31% percent showed ≥50% decrease in bone marrow blasts from baseline. Patients achieving an OR had a significant improvement in median progression-free survival (PFS) (5.1 vs 1.3 months; P = .008; hazard ratio [HR], 3.1) and overall survival (9.7 vs 2.7 months; P = .01; HR, 3.1) compared with nonresponders. These findings suggest that selinexor is safe as a monotherapy in patients with relapsed or refractory AML and have informed subsequent phase 2 clinical development. This trial was registered at www.clinicaltrials.gov as #NCT01607892.


Cancers ◽  
2021 ◽  
Vol 13 (16) ◽  
pp. 4113
Author(s):  
Fatih M. Uckun ◽  
Tara L. Lin ◽  
Alice S. Mims ◽  
Prapti Patel ◽  
Cynthia Lee ◽  
...  

APVO436 is a recombinant T cell-engaging humanized bispecific antibody designed to redirect host T cell cytotoxicity in an MHC-independent manner to CD123-expressing blast cells from patients with hematologic malignancies and has exhibited single-agent anti-leukemia activity in murine xenograft models of acute myeloid leukemia (AML). In this first-in-human (FIH) multicenter phase 1B study, we sought to determine the safety and tolerability of APVO436 in R/R AML/myelodysplastic syndrome (MDS) patients and identify a clinically active recommended phase 2 dose (RP2D) level for its further clinical development. A total of 46 R/R AML/MDS patients who had failed 1–8 prior lines of therapy received APVO436 as weekly intravenous (IV) infusions at 10 different dose levels, ranging from a Minimum Anticipated Biological Effect Level (MABEL) of 0.3 mcg to 60 mcg. APVO436 exhibited a favorable safety profile with acceptable tolerability and manageable drug-related adverse events (AEs), and its maximum tolerated dose (MTD) was not reached at a weekly dose of 60 mcg. The most common APVO436-related AEs were infusion-related reactions (IRR) occurring in 13 (28.3%) patients and cytokine release syndrome (CRS) occurring in 10 (21.7%). The single dose RP2D level was identified as 0.2 mcg/kg. Preliminary efficacy signals were observed in both AML and MDS patients: Prolonged stable disease (SD), partial remissions (PR), and complete remissions (CR) were observed in R/R AML patients as best overall responses to APVO436 at the RP2D level. Three of six evaluable MDS patients had marrow CRs. The safety and preliminary evidence of efficacy of APVO436 in R/R AML and MDS patients warrant further investigation of its clinical impact potential.


2020 ◽  
Vol 21 (14) ◽  
pp. 5135
Author(s):  
Edit Kotogány ◽  
József Á. Balog ◽  
Lajos I. Nagy ◽  
Róbert Alföldi ◽  
Valeria Bertagnolo ◽  
...  

Chemotherapy-induced differentiation of immature myeloid progenitors, such as acute myeloid leukemia (AML) cells or myeloid-derived suppressor cells (MDSCs), has remained a challenge for the clinicians. Testing our imidazo[1,2-b]pyrazole-7-carboxamide derivative on HL-60 cells, we obtained ERK phosphorylation as an early survival response to treatment followed by the increase of the percentage of the Bcl-xlbright and pAktbright cells. Following the induction of Vav1 and the AP-1 complex, a driver of cellular differentiation, FOS, JUN, JUNB, and JUND were elevated on a concentration and time-dependent manner. As a proof of granulocytic differentiation, the cells remained non-adherent, the expression of CD33 decreased; the granularity, CD11b expression, and MPO activity of HL-60 cells increased upon treatment. Finally, viability of HL-60 cells was hampered shown by the depolarization of mitochondria, activation of caspase-3, cleavage of Z-DEVD-aLUC, appearance of the sub-G1 population, and the leakage of the lactate-dehydrogenase into the supernatant. We confirmed the differentiating effect of our drug candidate on human patient-derived AML cells shown by the increase of CD11b and decrease of CD33+, CD7+, CD206+, and CD38bright cells followed apoptosis (IC50: 80 nM) after treatment ex vivo. Our compound reduced both CD11b+/Ly6C+ and CD11b+/Ly6G+ splenic MDSCs from the murine 4T1 breast cancer model ex vivo.


Sign in / Sign up

Export Citation Format

Share Document